1
|
Plata VTG, de Jesus Simão J, de Sousa AFB, Alonso-Vale MI, Armelin-Correa L. Impact of fish oil on epigenetic regulation in perirenal adipose tissue of obese mice. Obes Res Clin Pract 2025:S1871-403X(25)00043-2. [PMID: 40246605 DOI: 10.1016/j.orcp.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025]
Abstract
It has been demonstrated that fish oil (FO), a source of omega-3 polyunsaturated fatty acids (n-3 PUFA), offers partial protection to mice from the adverse effects of a high-fat diet (HFD) by altering the expression of genes involved in adipogenesis and adipocyte metabolism. Histone 3 lysine 27 (H3K27) modifiers, namely Ezh2, Kdm6a, Kdm6b, Crebbp and Ep300, are vital for the appropriate differentiation and metabolism of adipocytes, as they can either silence or activate transcription. The expansion of perirenal adipose tissue (AT) in obesity is associated with a number of complications, including hypertension and kidney disease. The aim of this study was to assess the expression of H3K27 modifiers and genes involved in adipogenesis and adipocyte metabolism in perirenal AT of HFD-fed and FO-treated (5DHA:1EPA) mice using real-time PCR. This study demonstrates, for the first time, that a high-fat diet (HFD) increases the expression of Kdm6b (H3K27 demethylase) in perirenal AT, and that treatment with FO can completely reverse this effect. Conversely, the expression of the Acly gene, which encodes an enzyme that provides a substrate for histone acetylases, was found to be reduced in HFD-fed mice and this was not reversed by FO treatment. Additionally, transcription factor genes, such as Tbx1, exhibited diminished expression in perirenal AT of mice fed an HFD. These observations suggest that a HFD affects the expression of chromatin modifiers, transcription factors, and metabolic genes in perirenal AT, and that FO can reverse some of these effects, offering a promising avenue for the treatment of obesity.
Collapse
Affiliation(s)
- Victor Tadeu Gonçalves Plata
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Jussara de Jesus Simão
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Andressa França Bispo de Sousa
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Maria Isabel Alonso-Vale
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil; Department of Biological Sciences, Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil
| | - Lucia Armelin-Correa
- Post-graduation Program in Chemical Biology Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil; Department of Biological Sciences, Institute of Environmental Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, Brazil.
| |
Collapse
|
2
|
Zhang M, Feng C, Zhang B, Yin Y, Chen J, Liu H, Farag MA, Mamadalieva NZ, Li N, Sun J, Sun S, Liu C. In vitro and in vivo immune-enhancing effects of punicic acid and the action mechanisms as revealed via microbiome and lipid profiling. Food Funct 2025; 16:3120-3133. [PMID: 40159912 DOI: 10.1039/d4fo05023a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Punicic acid (PA) is a chief component of pomegranate seed oil with several health benefits. In this study, the in vitro immunomodulatory activity of PA was assessed using RAW264.7 cells, revealing that PA activated the macrophages, facilitated the concentration of immune-related cytokines and enzymes, and regulated the immune-related NF-κB and MAPK signaling pathways. Further, the in vivo immune-enhancing effect of PA was evaluated with the cyclophosphamide (CTX)-induced immune-compromised mouse model with 16S rDNA amplicon sequencing and relative quantification of lipidome. Results indicated that high doses of PA (200 mg kg-1) remarkably restored CTX-induced immune injury by enhancing the innate and adaptive immunity to stimulate the secretion of immune-related factors. In addition, PA improved gut microbiota dysbiosis and ameliorated lipid metabolism disorders. Our research provides a theoretical basis for the exploitation of PA as a functional component with immune-enhancing effects and adds to the potential health uses of pomegranate seed oil.
Collapse
Affiliation(s)
- Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Caiyun Feng
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China
| | - Bo Zhang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China
| | - Yanlei Yin
- Shandong Institute of Pomology, Tai'an, 271000, China
| | - Jinlong Chen
- Work Station of Forest Fruit Industry in Kashi, Kashi, 844000, PR China
| | - Haoran Liu
- JiMei One Health Industry (Shandong) Co., Ltd, Zaozhuang, 277300, PR China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nilufar Z Mamadalieva
- Institute of the Chemistry of Plant Substances of the Academy Sciences of Uzbekistan, Tashkent, 100170, Uzbekistan
| | - Ningyang Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, PR China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Shutao Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, 23788 Gongye North Road, Jinan, 250100, PR China.
- Shandong Aojing Biotechnology Co., Ltd, Jining, 273500, PR China
| |
Collapse
|
3
|
Mercado-López L, Muñoz Y, Farias C, Beyer MP, Carrasco-Gutiérrez R, Caicedo-Paz AV, Dagnino-Subiabre A, Espinosa A, Valenzuela R. High-Fat Diet in Perinatal Period Promotes Liver Steatosis and Low Desaturation Capacity of Polyunsaturated Fatty Acids in Dams: A Link with Anxiety-Like Behavior in Rats. Nutrients 2025; 17:1180. [PMID: 40218938 PMCID: PMC11990584 DOI: 10.3390/nu17071180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study investigates the effects of a high-fat diet (HFD) during pregnancy and lactation on maternal and offspring health, focusing on behavioral, metabolic, and fatty acid composition outcomes in a rat model. METHODS Twelve female Sprague-Dawley rats were fed either a control diet, CD (n = 6), or HFD (n = 6) for 12 weeks, encompassing mating, gestation, and lactation periods (18 weeks). Anxiety-like behavior, maternal behavior, depression-like behavior, and social play were studied. Post mortem, the liver function, hepatic steatosis, and fatty acid composition (erythrocytes, liver, adipose tissue) were evaluated. In regard to desaturase enzymes (Δ-6D and Δ-5D), liver activity, protein mass, and gene expression (RT-PCR) were analyzed. Additionally, gene expression of PPAR-α, ACOX, CPT1-α, SREBP-1c, ACC, and FAS was assessed. Statistical analysis was performed using Student's t-test, mean ± SD (p < 0.05). RESULTS The HFD significantly increased maternal weight and anxiety-like behavior while reducing social interactions exclusively in male offspring (p < 0.05). It also led to a significant decrease in the synthesis and content of n-3 PUFAs in the analyzed tissues, induced hepatic steatosis, and upregulated the expression of pro-lipogenic genes in the maternal liver. CONCLUSIONS These findings suggest that long-term HFD consumption alters tissue fatty acid composition, disrupts metabolic homeostasis, and contributes to behavioral changes, increasing anxiety-like behaviors in pregnant dams and reducing social interactions in male offspring. Overall, this study provides further insight into the detrimental effects of HFD consumption during the perinatal period.
Collapse
Affiliation(s)
- Lorena Mercado-López
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
- Direccion de Postgrado, Facultad Medicina, Universidad Andres Bello, Santiago 8370035, Chile
| | - Yasna Muñoz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
- Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
| | - María Paz Beyer
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
- Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Robinson Carrasco-Gutiérrez
- Laboratory of Stress Neurobiology, Interdisciplinary Centre for Health Studies (CIESAL), Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Angie Vanessa Caicedo-Paz
- Escuela de Agronomía, Facultad de Ciencias Agronómicas y de los Alimentos, Pontificia Universidad Católica de Valparaíso, Quillota 2260000, Chile;
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Interdisciplinary Centre for Health Studies (CIESAL), Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Alejandra Espinosa
- Escuela de Medicina, Campus San Felipe, Universidad de Valparaíso, San Felipe 2170000, Chile;
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
| |
Collapse
|
4
|
Zhang B, Han Y, Cheng M, Yan L, Gao K, Zhou D, Wang A, Lin P, Jin Y. Metabolomic effects of intrauterine meloxicam perfusion on histotroph in dairy heifers during diestrus. Front Vet Sci 2025; 12:1528530. [PMID: 40171410 PMCID: PMC11959509 DOI: 10.3389/fvets.2025.1528530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
In ruminants, conceptus elongation is a crucial developmental process that depends on uterine lumen fluid (ULF) and coincides with a period of high pregnancy loss. Prostaglandins (PGs) play indispensable roles in conceptus elongation and implantation. However, the effects of uterus-derived PGs on the uterine environment remain unclear. To explore the metabolic pathways and metabolites induced by endometrium-derived PGs that may affect conceptus elongation and implantation in dairy cows, we investigated the biochemical composition of ULF following intrauterine perfusion of meloxicam from days 12 to 14 of the estrous cycle. Intrauterine administration of meloxicam significantly downregulated the prostaglandin-related metabolites in the ULF. A total of 385 distinct metabolites, primarily clustered within lipids and lipid-like molecules, organic acids and derivatives, organoheterocyclic compounds, and benzenoids, were identified. The metabolite network analysis identified 10 core metabolites as follows: S-adenosylhomocysteine, guanosine, inosine, thymidine, cholic acid, xanthine, niacinamide, prostaglandin I2, 5-hydroxyindoleacetic acid, and indoleacetaldehyde. The pathway enrichment analysis revealed three significantly altered metabolic pathways: arachidonic acid metabolism, tryptophan (Trp) metabolism, and linoleic acid metabolism. A total of five metabolites-guanosine, inosine, thymidine, butyryl-l-carnitine, and l-carnitine-were associated with attachment and pregnancy loss and could serve as predictors of fertility. This global metabolic study of ULF enhances our understanding of histotroph alternations induced by uterus-derived PGs during diestrus in dairy cows, with implications for improving dairy cow fertility.
Collapse
Affiliation(s)
- Beibei Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Han
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ming Cheng
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Longgang Yan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Kangkang Gao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Dong Zhou
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
5
|
Sheng ZH, Gong XY, Huang PP, Xu QY, Zhang WJ, Sun FB, Song KY, Zeng DC. An Innovative Anoikis Signature With Inflammatory Infiltrates in Osteoarthritis. Int J Rheum Dis 2025; 28:e70093. [PMID: 39895467 DOI: 10.1111/1756-185x.70093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/15/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
AIM To explore the relationship between an innovative anoikis-related gene signature and inflammatory infiltrates in patients with osteoarthritis. METHODS Gene expression profiles (GSM1248759 and GSE200843) were curated from the Gene Expression Omnibus database, followed by the construction of a protein-protein interaction network. Functional and genomic enrichment analyses were conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. The CIBERSORT method was employed to investigate immune cell infiltration differences between osteoarthritic and control tissues. Additionally, the ConsensusClusterPlus package in R software was utilized to identify distinct anoikis patterns (Cluster C1 and Cluster C2) and conduct molecular biological investigations. RESULTS Analysis revealed two distinct anoikis patterns (Cluster C1 and Cluster C2), with Cluster C2 patients exhibiting varying immune cell levels compared to Cluster C1 patients. Molecular investigations identified 84 DEGs enriched in specific pathways such as adipocytokine signaling, cytokine-cytokine receptor interaction, ECM-receptor interaction, and the PPAR signaling pathway. qPCR experiments confirmed the elevated expression levels of specific genes, including SOD2, MAPK14, CEACM3, LAMB3, COL13A1, TLR3, NOTCH3, and KLF12, in the IL-1β-induced group compared with the osteoarthritis group. CONCLUSION This study highlights the role of anoikis-related genes and immune infiltration differences in osteoarthritis, enhancing our understanding of its development.
Collapse
Affiliation(s)
- Ze-Hao Sheng
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xin-Yi Gong
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peng-Peng Huang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi-Yu Xu
- Department of Rehabilitation Medicine, Anhui no.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Wen-Jie Zhang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng-Bao Sun
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kai-Yi Song
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Du-Chun Zeng
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Feng W, He Q, Li J, Zhou J, Hua G, Xu Y, Jiang G, Tang Y. Variability in Morphological Traits and Nutritional Profiles of Adult Eriocheir sinensis in Different Aquacultural Regions. Animals (Basel) 2025; 15:243. [PMID: 39858243 PMCID: PMC11759138 DOI: 10.3390/ani15020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Eriocheir sinensis is a species of significant economic importance in China's aquaculture industry. In order to investigate the variations in morphologic and nutritional profiles of farmed crabs across diverse farming regions, E. sinensis were collected from six aquafarms located in the primary cultivation areas in China. The findings revealed significant variations in specific morphological indices, but these are insufficient for clear regional distinction using PCA. Essential amino acid (EAA) levels varied by tissue and gender, with ovaries from the YY (Yongyan in Anhui province) group showing the highest levels, while the hepatopancreas in females from the JX (Jianxian in Jiangxi province) group had the lowest. The analysis of fatty acid contents revealed that muscles were rich in PUFAs, whereas MUFAs were dominant in the hepatopancreas and gonads. The fatty acid levels were significantly higher in ovaries than in testes, notably in the HZ (Huzhou in Zhejiang province), EZ (Ezhou in Hubei province), YY, and JX groups. Furthermore, the PCA based on proximate composition, amino acids, and fatty acids revealed distinct categorizations of E. sinensis from various regions. In conclusion, these findings highlight significant regional variations in the morphology and nutritional composition of farmed E. sinensis, suggesting the need for tailored aquaculture practices to optimize quality and output.
Collapse
Affiliation(s)
- Wenrong Feng
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (W.F.); (Q.H.); (J.L.); (Y.X.); (G.J.)
| | - Qinghong He
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (W.F.); (Q.H.); (J.L.); (Y.X.); (G.J.)
| | - Jianlin Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (W.F.); (Q.H.); (J.L.); (Y.X.); (G.J.)
| | - Jun Zhou
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing 210017, China;
| | - Guoan Hua
- China Jiangsu Haorun Biological Industry Group Co., Ltd., Taizhou 225311, China;
| | - Yuanfeng Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (W.F.); (Q.H.); (J.L.); (Y.X.); (G.J.)
| | - Gang Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (W.F.); (Q.H.); (J.L.); (Y.X.); (G.J.)
| | - Yongkai Tang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (W.F.); (Q.H.); (J.L.); (Y.X.); (G.J.)
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| |
Collapse
|
7
|
Zhang Y, Liu Y, Zhang M, Li G, Zhu S, Xie K, Xiao B, Li L. Clinical Relevance and Drug Modulation of PPAR Signaling Pathway in Triple-Negative Breast Cancer: A Comprehensive Analysis. PPAR Res 2024; 2024:4164906. [PMID: 39735727 PMCID: PMC11681981 DOI: 10.1155/ppar/4164906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is highly heterogeneous and poses a significant medical challenge due to limited treatment options and poor outcomes. Peroxisome proliferator-activated receptors (PPARs) play a crucial role in regulating metabolism and cell fate. While the association between PPAR signal and human cancers has been a topic of concern, its specific relationship with TNBC remains unclear. Integrated analysis of large published datasets from clinical cohorts and cell lines through databases has proven to be a powerful and essential approach for understanding cancer and uncovering new molecular targets. Here, we conducted a comprehensive study investigating the clinical relevance and drug modulation of the PPAR signaling pathway in TNBC, using data from The Cancer Genome Atlas (TCGA) for TNBC patients and Genomics of Drug Sensitivity in Cancer (GDSC) for TNBC cell lines, along with drug perturbation information from Connectivity Map (CMap). In the TCGA-TNBC cohort, higher PPAR signaling activity was not associated with clinical stage, prognosis, tumor mutational burden, microsatellite instability, homologous recombination deficiency, stemness, or proliferation status. However, it was linked to older age; an elevated rate of piccolo presynaptic cytomatrix protein (PCLO) mutations; and oncogenic signal transduction involving MAPK, Ras, and PI3K-Akt pathways. Additionally, it influenced biological pathways including fatty acid metabolism, AMPK signaling, and ferroptosis. Strikingly, higher PPAR activity appeared to promote the formation of an antitumor immune and microbial microenvironment. In the GDSC-TNBC cells, nevertheless, it seemed to incur chemoresistance. Furthermore, we identified a batch of potential compounds that can regulate the PPAR signaling pathway. Lastly, our experimental validation demonstrated the ability of the histone deacetylase (HDAC) inhibitor chidamide to activate the PPAR signal in TNBC cells. In conclusion, the PPAR signaling pathway likely has pleiotropic biological effects in TNBC. These preliminary but interesting findings enhance our understanding of the role played by PPAR signal and provide new insights into the heterogeneity driven by it in TNBC.
Collapse
Affiliation(s)
- Yanxia Zhang
- Department of Laboratory Medicine, The Sixth School of Clinical Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- School of Medicine, The South China University of Technology, Guangzhou, China
| | - Yunduo Liu
- Department of Laboratory Medicine, The Sixth School of Clinical Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- School of Public Health, Dali University, Dali, China
| | - Mei Zhang
- Department of Laboratory Medicine, The Sixth School of Clinical Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Guanjie Li
- Thyroid and Breast Specialty of General Surgery Area Five, The Sixth School of Clinical Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Siling Zhu
- Department of Laboratory Medicine, The Sixth School of Clinical Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Keping Xie
- School of Medicine, The South China University of Technology, Guangzhou, China
| | - Bin Xiao
- Department of Laboratory Medicine, Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, China
- The Fifth College of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth School of Clinical Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
8
|
Esmaili H, Tajik B, Tuomainen TP, Kurl S, Salonen JT, Virtanen JK. Associations of the serum n-6 PUFA concentrations with exercise-induced myocardial ischaemia in middle-aged and older men. Br J Nutr 2024; 132:1522-1529. [PMID: 39523844 DOI: 10.1017/s0007114524001971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
n-6 PUFA, especially linoleic acid (LA) but also arachidonic acid (AA), have been inversely associated with CHD. However, mechanisms underlying these associations are not fully known. We investigated the associations of the serum concentrations of total n-6 PUFA, LA, AA, γ-linolenic acid (GLA) and dihomo-γ-linolenic acid (DGLA), with the odds of myocardial ischaemia during exercise, a predictor of future cardiac events. A total of 1871 men without a history of CHD from the Kuopio Ischaemic Heart Disease Risk Factor Study (KIHD) aged 42-60 years were included. All participants performed a maximal symptom-limited exercise stress test, using an electrically braked bicycle ergometer. Multivariable-adjusted logistic regression was used to assess the OR for exercise-induced myocardial ischaemia in quartiles of the serum n-6 PUFA concentrations. After multivariable adjustment, men in the highest v. the lowest serum AA concentration had 50 % lower odds for exercise-induced myocardial ischaemia (OR 0·50, 95 % CI 0·34, 0·76; P-trend across quartiles < 0·001). For the other PUFA, the OR (95 % CI) were 1·00 (0·69, 1·46; P-trend = 0·89) for LA, 1·07 (0·75, 1·53; P-trend = 0·40) for GLA and 0·74 (0·51, 1·07; P-trend = 0·16) for DGLA. Among the n-6 PUFA, higher serum concentration of AA was associated with lower odds for myocardial ischaemia during an exercise test in middle-aged and older men. This may provide one mechanism for the previously observed possible cardioprotective properties of AA. Our findings also suggest that n-6 PUFA should not be considered as one homogenous group.
Collapse
Affiliation(s)
- Haleh Esmaili
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Behnam Tajik
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Tomi-Pekka Tuomainen
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Sudhir Kurl
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Jukka T Salonen
- University of Helsinki, The Faculty of Medicine, Department of Public Health, Helsinki, Finland
- MAS-Metabolic Analytical Services Oy, Helsinki, Finland
| | - Jyrki K Virtanen
- University of Eastern Finland, Kuopio Campus, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| |
Collapse
|
9
|
Loix M, Vanherle S, Turri M, Kemp S, Fernandes KJL, Hendriks JJA, Bogie JFJ. Stearoyl-CoA desaturase-1: a potential therapeutic target for neurological disorders. Mol Neurodegener 2024; 19:85. [PMID: 39563397 PMCID: PMC11575020 DOI: 10.1186/s13024-024-00778-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
Disturbances in the fatty acid lipidome are increasingly recognized as key drivers in the progression of various brain disorders. In this review article, we delve into the impact of Δ9 fatty acid desaturases, with a particular focus on stearoyl-CoA desaturase-1 (SCD1), within the setting of neuroinflammation, neurodegeneration, and brain repair. Over the past years, it was established that inhibition or deficiency of SCD1 not only suppresses neuroinflammation but also protects against neurodegeneration in conditions such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease. This protective effect is achieved through different mechanisms including enhanced remyelination, reversal of synaptic and cognitive impairments, and mitigation of α-synuclein toxicity. Intriguingly, metabolic rerouting of fatty acids via SCD1 improves the pathology associated with X-linked adrenoleukodystrophy, suggesting context-dependent benign and harmful effects of SCD1 inhibition in the brain. Here, we summarize and discuss the cellular and molecular mechanisms underlying both the beneficial and detrimental effects of SCD1 in these neurological disorders. We explore commonalities and distinctions, shedding light on potential therapeutic challenges. Additionally, we touch upon future research directions that promise to deepen our understanding of SCD1 biology in brain disorders and potentially enhance the clinical utility of SCD1 inhibitors.
Collapse
Affiliation(s)
- Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Marta Turri
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, NH, Netherlands
| | - Karl J L Fernandes
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
- University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
10
|
Zhuang P, Ao Y, Liu X, Ye H, Li H, Wan X, Zhang Y, Jiao J. Circulating fatty acids and risk of severe non-alcoholic fatty liver disease in the UK biobank: a prospective cohort of 116 223 individuals. Food Funct 2024; 15:10527-10538. [PMID: 39370886 DOI: 10.1039/d4fo01182a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Fatty acid (FA) metabolism plays an important role in the development of nonalcoholic fatty liver disease (NAFLD). However, data on the relationship between circulating FAs and NAFLD risk are limited. This study aims to assess the associations between specific circulating FAs and severe NAFLD risk among the general population. Overall 116 223 participants without NAFLD and other liver diseases from the UK Biobank were enrolled between 2006 and 2010 and were followed up until the end of 2021. Plasma concentrations of saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs), and polyunsaturated fatty acids (PUFAs) were analyzed using an NMR-based biomarker profiling platform. Hazard ratios (HRs) and 95% confidence intervals (CIs) of NAFLD risk were estimated using Cox proportional-hazard models adjusted for other potential confounders. During a mean follow-up of 12.3 years, we documented 1394 cases of severe NAFLD. After multivariate adjustment, plasma SFAs and MUFAs were associated with a higher risk of severe NAFLD, whereas plasma n-3 PUFAs, n-6 PUFAs, and linoleic acid (LA) were associated with a lower risk. As compared with the lowest quartile, HRs (95% CIs) of severe NAFLD risk in the highest quartiles were 1.85 (1.45-2.36) for SFAs, 1.74 (1.23-2.44) for MUFAs, 0.79 (0.65-0.97) for n-3 PUFAs, 0.68 (0.48-0.96) for n-6 PUFAs, and 0.73 (0.54-0.99) for LA. The significant relationships were mainly mediated by serum TG for SFAs, HDL-C for MUFAs and n-6 PUFAs, and C-reactive protein for n-3 PUFAs. Plasma SFAs were associated with a more pronounced increase in the risk of severe NAFLD among participants with fewer SFA-associated alleles (P interaction = 0.032). Dietary recommendations for reducing plasma SFAs and MUFAs while increasing n-3 and n-6 PUFAs may be protective for severe NAFLD, which could be mediated by lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Yang Ao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Hao Ye
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Haoyu Li
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xuzhi Wan
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
11
|
Pinheiro CG, Motta BP, Oliveira JO, Cardoso FN, Figueiredo ID, Machado RTA, da Silva PB, Chorilli M, Brunetti IL, Baviera AM. Bixin Combined with Metformin Ameliorates Insulin Resistance and Antioxidant Defenses in Obese Mice. Pharmaceuticals (Basel) 2024; 17:1202. [PMID: 39338363 PMCID: PMC11434661 DOI: 10.3390/ph17091202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Bixin (C25H30O4; 394.51 g/mol) is the main apocarotenoid found in annatto seeds. It has a 25-carbon open chain structure with a methyl ester group and carboxylic acid. Bixin increases the expression of antioxidant enzymes, which may be interesting for counteracting oxidative stress. This study investigated whether bixin-rich annatto extract combined with metformin was able to improve the disturbances observed in high-fat diet (HFD)-induced obesity in mice, with an emphasis on markers of oxidative damage and antioxidant defenses. HFD-fed mice were treated for 8 weeks with metformin (50 mg/kg) plus bixin-rich annatto extract (5.5 and 11 mg/kg). This study assessed glucose tolerance, insulin sensitivity, lipid profile and paraoxonase 1 (PON-1) activity in plasma, fluorescent AGEs (advanced glycation end products), TBARSs (thiobarbituric acid-reactive substances), and the activities of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) in the liver and kidneys. Treatment with bixin plus metformin decreased body weight gain, improved insulin sensitivity, and decreased AGEs and TBARSs in the plasma, liver, and kidneys. Bixin plus metformin increased the activities of PON-1, SOD, CAT, and GSH-Px. Bixin combined with metformin improved the endogenous antioxidant defenses in the obese mice, showing that this combined therapy may have the potential to contrast the metabolic complications resulting from oxidative stress.
Collapse
Affiliation(s)
- Camila Graça Pinheiro
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| | - Bruno Pereira Motta
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| | - Juliana Oriel Oliveira
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| | - Felipe Nunes Cardoso
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| | - Ingrid Delbone Figueiredo
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| | - Rachel Temperani Amaral Machado
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (R.T.A.M.); (P.B.d.S.); (M.C.)
| | - Patrícia Bento da Silva
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (R.T.A.M.); (P.B.d.S.); (M.C.)
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (R.T.A.M.); (P.B.d.S.); (M.C.)
| | - Iguatemy Lourenço Brunetti
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| | - Amanda Martins Baviera
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (C.G.P.); (B.P.M.); (J.O.O.); (F.N.C.); (I.D.F.); (I.L.B.)
| |
Collapse
|
12
|
Yang X, Li X, Hu M, Huang J, Yu S, Zeng H, Mao L. EPA and DHA differentially improve insulin resistance by reducing adipose tissue inflammation-targeting GPR120/PPARγ pathway. J Nutr Biochem 2024; 130:109648. [PMID: 38631512 DOI: 10.1016/j.jnutbio.2024.109648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
Insulin resistance (IR) is a global health challenge, often initiated by dysfunctional adipose tissue. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may have different effects on IR, but the mechanisms are unknown. This study aims to evaluate the protective effect of EPA and DHA against IR in a high-fat diet (HFD) mice model and investigate whether EPA and DHA alter IR modulate the G-protein-poupled receptor 120/peroxisome proliferator-activated receptor γ (GPR120/PPARγ) pathway in macrophages and adipocytes, which may affect IR in adipocytes. The findings of this study show that 4% DHA had a better effect in improving IR and reducing inflammatory cytokines in adipose tissue of mice. Additionally, in the cell experiment, the use of AH7614 (a GPR120 antagonist) inhibited the glucose consumption increase and the increasable expression of PPARγ and insulin signaling molecules mediated by DHA in adipocytes. Furthermore, GW9662 (a PPARγ antagonist) hindered the upregulation of glucose consumption and insulin signaling molecule expression induced by EPA and DHA in adipocytes. DHA exhibited significant effects in reducing the number of migrated cells and inflammation. The compounds AH7614 and GW9662 hindered the suppressive effects of EPA and DHA on macrophage-induced IR in adipocytes. These findings suggest that DHA has a stronger potential in improving IR in adipocytes through the GPR120/PPARγ pathway in macrophages, when compared to EPA.
Collapse
Affiliation(s)
- Xian Yang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xudong Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Manjiang Hu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jie Huang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Siyan Yu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Huanting Zeng
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Limei Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
13
|
Kaimala S, Lootah SS, Mehra N, Kumar CA, Marzooqi SA, Sampath P, Ansari SA, Emerald BS. The Long Non-Coding RNA Obesity-Related (Obr) Contributes To Lipid Metabolism Through Epigenetic Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401939. [PMID: 38704700 PMCID: PMC11234455 DOI: 10.1002/advs.202401939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Indexed: 05/07/2024]
Abstract
Obesity is a multifactorial disease that is part of today's epidemic and also increases the risk of other metabolic diseases. Long noncoding RNAs (lncRNAs) provide one tier of regulatory mechanisms to maintain metabolic homeostasis. Although lncRNAs are a significant constituent of the mammalian genome, studies aimed at their metabolic significance, including obesity, are only beginning to be addressed. Here, a developmentally regulated lncRNA, termed as obesity related (Obr), whose expression in metabolically relevant tissues such as skeletal muscle, liver, and pancreas is altered in diet-induced obesity, is identified. The Clone 9 cell line and high-fat diet-induced obese Wistar rats are used as a model system to verify the function of Obr. By using stable expression and antisense oligonucleotide-mediated downregulation of the expression of Obr followed by different molecular biology experiments, its role in lipid metabolism is verified. It is shown that Obr associates with the cAMP response element-binding protein (Creb) and activates different transcription factors involved in lipid metabolism. Its association with the Creb histone acetyltransferase complex, which includes the cAMP response element-binding protein (CBP) and p300, positively regulates the transcription of genes involved in lipid metabolism. In addition, Obr is regulated by Pparγ in response to lipid accumulation.
Collapse
Affiliation(s)
- Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Shareena Saeed Lootah
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Neha Mehra
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Challagandla Anil Kumar
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Saeeda Al Marzooqi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| | - Prabha Sampath
- A*STAR Skin Research Laboratory, Agency for Science Technology & Research (A*STAR), Singapore, 138648, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Genome Institute of Singapore, Agency for Science Technology & Research (A*STAR), Singapore, 138672, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| |
Collapse
|
14
|
Lu C, Zhou X, Zhang L. Phospholipids with two polyunsaturated fatty acyl tails: an important driver of ferroptosis. MedComm (Beijing) 2024; 5:e606. [PMID: 38919333 PMCID: PMC11196922 DOI: 10.1002/mco2.606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/27/2024] Open
Abstract
We highlight the latest work of Qiu et al. on the core mechanism of ferroptosis induced by rare phospholipids with two polyunsaturated fatty acyl tails (PL-PUFA2s), which has been published in Cell. It has long been acknowledged that PLs containing one PUFA tail (PL-PUFA1s) serve as substrates for phospholipid peroxidation during the process of ferroptosis, owing to their susceptibility to oxidation and prevalence in vivo. However, the authors note that PL-PUFA2s, rather than PL-PUFA1s, represent critical lipid classes involved in the pro-ferroptosis process. Exogenous phosphatidylcholine-PUFA2s accumulate in mitochondria and combine with Complex I within the electron transport chain, thereby potentially resulting in an elevation of mitochondrial reactive oxygen species levels. Then, these mitochondrial peroxides prompt the substantial accumulation of peroxides within the endoplasmic reticulum, ultimately culminating in ferroptosis. These findings shed light on the potential molecular mechanisms underlying the induction of ferroptosis by dietary PL-PUFA2s and offer novel insights for both the evaluation of cellular iron death sensitivity and the treatment of cancer. This article will provide a more comprehensive elucidation of the paper and facilitate an enhanced understanding of the underlying mechanisms for readers.
Collapse
Affiliation(s)
- Chu Lu
- Life Sciences Institute, The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyThe Second Affiliated Hospital of the Zhejiang University School of MedicineZhejiang UniversityHangzhouChina
| | - Xiaoxue Zhou
- Life Sciences Institute, The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyThe Second Affiliated Hospital of the Zhejiang University School of MedicineZhejiang UniversityHangzhouChina
| | - Long Zhang
- Life Sciences Institute, The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell BiologyThe Second Affiliated Hospital of the Zhejiang University School of MedicineZhejiang UniversityHangzhouChina
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid TumorsThe First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- Cancer Center Zhejiang UniversityHangzhouChina
| |
Collapse
|
15
|
Ruswandi YAR, Lesmana R, Rosdianto AM, Gunadi JW, Goenawan H, Zulhendri F. Understanding the Roles of Selenium on Thyroid Hormone-Induced Thermogenesis in Adipose Tissue. Biol Trace Elem Res 2024; 202:2419-2441. [PMID: 37758980 DOI: 10.1007/s12011-023-03854-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Brown adipose tissue (BAT) and white adipose tissue (WAT) are known to regulate lipid metabolism. A lower amount of BAT compared to WAT, along with adipose tissue dysfunction, can result in obesity. Studies have shown that selenium supplementation protects against adipocyte dysfunction, decreases WAT triglycerides, and increases BAT triiodothyronine (T3). In this review, we discuss the relationship between selenium and lipid metabolism regulation through selenoprotein deiodinases and the role of deiodinases and thyroid hormones in the induction of adipose tissue thermogenesis. Upon 22 studies included in our review, we found that studies investigating the relationship between selenium and deiodinases demonstrated that selenium supplementation affects the iodothyronine deiodinase 2 (DIO2) protein and the expression of its associated gene, DIO2, proportionally. However, its effect on DIO1 is inconsistent while its effect on DIO3 activity is not detected. Studies have shown that the activity of deiodinases especially DIO2 protein and DIO2 gene expression is increased along with other browning markers upon white adipose tissue browning induction. Studies showed that thermogenesis is stimulated by the thyroid hormone T3 as its activity is correlated to the expression of other thermogenesis markers. A proposed mechanism of thermogenesis induction in selenium supplementation is by autophagy control. However, more studies are needed to establish the role of T3 and autophagy in adipose tissue thermogenesis, especially, since some studies have shown that thermogenesis can function even when T3 activity is lacking and studies related to autophagy in adipose tissue thermogenesis have contradictory results.
Collapse
Affiliation(s)
- Yasmin Anissa R Ruswandi
- Graduate School of Master Program in Anti-Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia.
| | - Aziiz Mardanarian Rosdianto
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia
- Veterinary Medicine Study Program, Faculty of Medicine, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Hanna Goenawan
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang, KM.21, Hegarmanah, Kec. Jatinangor, Kabupaten Sumedang, West Java, 45363, Indonesia
| | - Felix Zulhendri
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Kabupaten Sumedang, West Java, Indonesia
- Kebun Efi, Kabanjahe, 22171, North Sumatra, Indonesia
| |
Collapse
|
16
|
Zhang T, Xie L, Guo Y, Wang Z, Guo X, Liu R, Jin Q, Chang M, Wang X. 4,4-Dimethylsterols Reduces Fat Accumulation via Inhibiting Fatty Acid Amide Hydrolase In Vitro and In Vivo. RESEARCH (WASHINGTON, D.C.) 2024; 7:0377. [PMID: 38812531 PMCID: PMC11134202 DOI: 10.34133/research.0377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/14/2024] [Indexed: 05/31/2024]
Abstract
4,4-Dimethylsterols constitute a unique class of phytosterols responsible for regulating endogenous cannabinoid system (ECS) functions. However, precise mechanism through which 4,4-dimethylsterols affect fat metabolism and the linkage to the ECS remain unresolved. In this study, we identified that 4,4-dimethylsterols, distinct from 4-demethseterols, act as inhibitors of fatty acid amide hydrolases (FAAHs) both in vivo and in vitro. Genetic ablation of FAAHs (faah-1) abolishes the effects of 4,4-dimethylsterols on fat accumulation and locomotion behavior in a Caenorhabditis elegans model. We confirmed that dietary intervention with 4,4-dimethylsterols in a high-fat diet (HFD) mouse model leads to a significant reduction in body weight (>11.28%) with improved lipid profiles in the liver and adipose tissues and increased fecal triacylglycerol excretion. Untargeted and targeted metabolomics further verified that 4,4-dimethylsterols influence unsaturated fatty acid biosynthesis and elevate oleoyl ethanolamine levels in the intestine. We propose a potential molecular mechanism in which 4,4-dimethylsterols engage in binding interactions with the catalytic pocket (Ser241) of FAAH-1 protein due to the shielded polarity, arising from the presence of 2 additional methyl groups (CH3). Consequently, 4,4-dimethylsterols represent an unexplored class of beneficial phytosterols that coordinate with FAAH-1 activity to reduce fat accumulation, which offers new insight into intervention strategies for treating diet-induced obesity.
Collapse
Affiliation(s)
- Tao Zhang
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology,
Jiangnan University, Wuxi 214122, China
- College of Food Science and Technology,
Huazhong Agricultural University, Wuhan 430070, China
| | - Liangliang Xie
- School of Biological and Food Engineering,
Anhui Polytechnic University, Wuhu 241000, China
| | - Yiwen Guo
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology,
Jiangnan University, Wuxi 214122, China
| | - Zhangtie Wang
- College of Biosystems Engineering and Food Science,
Zhejiang University, Hangzhou 310058, China
| | - Xin Guo
- Department of Food Science,
University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ruijie Liu
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology,
Jiangnan University, Wuxi 214122, China
| | - Qingzhe Jin
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology,
Jiangnan University, Wuxi 214122, China
| | - Ming Chang
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology,
Jiangnan University, Wuxi 214122, China
| | - Xingguo Wang
- School of Food Science and Technology, State Key Laboratory of Food Science and Technology,
Jiangnan University, Wuxi 214122, China
| |
Collapse
|
17
|
Vasques-Monteiro IML, Fernandes-da-Silva A, Miranda CS, Silva-Veiga FM, Daleprane JB, Souza-Mello V. Anti-steatotic effects of PPAR-alpha and gamma involve gut-liver axis modulation in high-fat diet-fed mice. Mol Cell Endocrinol 2024; 585:112177. [PMID: 38373652 DOI: 10.1016/j.mce.2024.112177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/21/2024]
Abstract
AIM To evaluate the effects of PPARα and PPARγ activation (alone or in combination) on the gut-liver axis, emphasizing the integrity of the intestinal barrier and hepatic steatosis in mice fed a high saturated fat diet. METHODS Male C57BL/6J were fed a control diet (C) or a high-fat diet (HF) for ten weeks. Then, a four-week treatment started: HF-α (WY14643), HF-γ (low-dose pioglitazone), and HF-αγ (combination). RESULTS The HF caused overweight, insulin resistance, impaired gut-liver axis, and marked hepatic steatosis. Treatments reduced body mass, improved glucose homeostasis, and restored the gut microbiota diversity and intestinal barrier gene expression. Treatments also lowered the plasma lipopolysaccharide concentrations and favored beta-oxidation genes, reducing macrophage infiltration and steatosis in the liver. CONCLUSION Treatment with PPAR agonists modulated the gut microbiota and rescued the integrity of the intestinal barrier, alleviating hepatic steatosis. These results show that these agonists can contribute to metabolic-associated fatty liver disease treatment.
Collapse
Affiliation(s)
- Isabela Macedo Lopes Vasques-Monteiro
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Aline Fernandes-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carolline Santos Miranda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Flavia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics (LEING), Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Papin M, Fontaine D, Goupille C, Figiel S, Domingo I, Pinault M, Guimaraes C, Guyon N, Cartron PF, Emond P, Lefevre A, Gueguinou M, Crottès D, Jaffrès PA, Ouldamer L, Maheo K, Fromont G, Potier-Cartereau M, Bougnoux P, Chantôme A, Vandier C. Endogenous ether lipids differentially promote tumor aggressiveness by regulating the SK3 channel. J Lipid Res 2024; 65:100544. [PMID: 38642894 PMCID: PMC11127165 DOI: 10.1016/j.jlr.2024.100544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/22/2024] Open
Abstract
SK3 channels are potassium channels found to promote tumor aggressiveness. We have previously demonstrated that SK3 is regulated by synthetic ether lipids, but the role of endogenous ether lipids is unknown. Here, we have studied the role of endogenous alkyl- and alkenyl-ether lipids on SK3 channels and on the biology of cancer cells. Experiments revealed that the suppression of alkylglycerone phosphate synthase or plasmanylethanolamine desaturase 1, which are key enzymes for alkyl- and alkenyl-ether-lipid synthesis, respectively, decreased SK3 expression by increasing micro RNA (miR)-499 and miR-208 expression, leading to a decrease in SK3-dependent calcium entry, cell migration, and matrix metalloproteinase 9-dependent cell adhesion and invasion. We identified several ether lipids that promoted SK3 expression and found a differential role of alkyl- and alkenyl-ether lipids on SK3 activity. The expressions of alkylglycerone phosphate synthase, SK3, and miR were associated in clinical samples emphasizing the clinical consistency of our observations. To our knowledge, this is the first report showing that ether lipids differentially control tumor aggressiveness by regulating an ion channel. This insight provides new possibilities for therapeutic interventions, offering clinicians an opportunity to manipulate ion channel dysfunction by adjusting the composition of ether lipids.
Collapse
Affiliation(s)
- Marion Papin
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Delphine Fontaine
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Caroline Goupille
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France; Department of Gynecology, CHRU Bretonneau, Tours, France
| | - Sandy Figiel
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Isabelle Domingo
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Michelle Pinault
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Cyrille Guimaraes
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Nina Guyon
- CRCINA-INSERM 1232, Equipe « Apoptose et Progression tumorale », Nantes, France
| | | | - Patrick Emond
- iBrain, UMR 1253, INSERM, Université de Tours, Tours, France; Nuclear medicine in vitro department, CHRU Bretonneau, Tours, France
| | - Antoine Lefevre
- iBrain, UMR 1253, INSERM, Université de Tours, Tours, France
| | - Maxime Gueguinou
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - David Crottès
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Paul-Alain Jaffrès
- Laboratoire Chimie Electrochimie Moléculaires et Chimie Analytique (CEMCA), UMR 6521, CNRS, University of Brest, Brest, France
| | - Lobna Ouldamer
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France; Department of Gynecology, CHRU Bretonneau, Tours, France
| | - Karine Maheo
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Gaëlle Fromont
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France; Department of Pathology, CHRU Bretonneau, Tours, France
| | - Marie Potier-Cartereau
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Philippe Bougnoux
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Aurélie Chantôme
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France
| | - Christophe Vandier
- Niche, Nutrition, Cancer & Oxidative metabolism (N2COx), UMR 1069, INSERM, University of Tours, Tours, France.
| |
Collapse
|
19
|
Valenzuela R, Walbaum B, Farias C, Acevedo F, Vargas C, Bennett JT, Bravo ML, Pinto MP, Medina L, Merino T, Ibañez C, Parada A, Sanchez C. High linoleic acid levels in red blood cells predict a poor response to neoadjuvant chemotherapy in human epidermal growth factor receptor type 2-positive breast cancer patients. Nutrition 2024; 121:112357. [PMID: 38430738 DOI: 10.1016/j.nut.2024.112357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Polyunsaturated fatty acids are categorized as ω-3 or ⍵-6. Previous studies demonstrate that breast cancers display a high expression of fatty acid synthase and high fatty acid levels. Our study sought to determine if changes in plasma or red blood cell membrane fatty acid levels were associated with the response to preoperative (neoadjuvant) chemotherapy in non-metastatic breast cancer patients. METHODS Our prospective study assessed fatty acid levels in plasma and red blood cell membrane. Response to neoadjuvant chemotherapy was evaluated by the presence or absence of pathologic complete response and/or residual cancer burden. RESULTS A total of 28 patients were included. First, patients who achieved pathologic complete response had significantly higher neutrophil-to-lymphocyte ratio versus no pathologic complete response (P = 0.003). Second, total red blood cell membrane polyunsaturated fatty acids were higher in the absence of pathologic complete response (P = 0.0028). Third, total red blood cell membrane ⍵-6 polyunsaturated fatty acids were also higher in no pathologic complete response (P < 0.01). Among ⍵-6 polyunsaturated fatty acids, red blood cell membrane linoleic acid was higher in the absence of pathologic complete response (P < 0.01). Notably, plasma polyunsaturated fatty acid, ⍵-6, and linoleic acid levels did not have significant differences. A multivariate analysis confirmed red blood cell membrane linoleic acid was associated with no pathologic complete response; this was further confirmed by receiver operating characteristic analysis (specificity = 92.3%, sensitivity = 76.9%, and area under the curve = 0.855). CONCLUSIONS Pending further validation, red blood cell membrane linoleic acid might serve as a predictor biomarker of poorer response to neoadjuvant chemotherapy in non-metastatic human epidermal growth factor receptor type 2-positive breast cancer. Measuring fatty acids in red blood cell membrane could offer a convenient, minimally invasive strategy to identifying patients more likely to respond or those with chemoresistance.
Collapse
Affiliation(s)
- Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Benjamín Walbaum
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Francisco Acevedo
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Vargas
- Department of Surgical Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Tomas Bennett
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Loreto Bravo
- Support Team for Oncological Research and Medicine (STORM), Santiago, Chile
| | - Mauricio P Pinto
- Support Team for Oncological Research and Medicine (STORM), Santiago, Chile
| | - Lidia Medina
- Centro del Cáncer Nuestra Señora de la Esperanza, UC CHRISTUS Healthcare Network, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tomas Merino
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Ibañez
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Parada
- Department of Health Sciences. School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cesar Sanchez
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
20
|
Muñoz Y, Mercado L, Farias C, Beyer MP, Alvear I, Echeverría F, Valenzuela R. Impact of polyunsaturated fatty acids during and pregnancy and lactation: A comprehensive review. Prostaglandins Leukot Essent Fatty Acids 2024; 203:102656. [PMID: 39481326 DOI: 10.1016/j.plefa.2024.102656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/17/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND Docosahexaenoic acid (DHA) and arachidonic acid (ARA) are relevant polyunsaturated fatty acids (PUFA) derived from alpha-linolenic acid (ALA) and linoleic acid (LA), respectively. These are important in pregnancy and lactation periods because of their benefits to the developing fetus and infant. Currently, the high prevalence of gestational obesity has led to a revision of PUFAs recommendations in these periods, due to changes in the lipid profile of women marked by a higher consumption of n-6 PUFA. OBJECTIVE This review aims to present an updated compilation of evidence on DHA and ARA during gestation and lactation. METHODS The literature review was performed in different databases, PubMed, Scopus, Web of Science, Scielo, ISI, and Ovid MedLine, highlighting the importance of DHA and ARA for newborn development. RESULTS An adequate intake of n-3 PUFA, especially DHA, in the mother during pregnancy and the postnatal period is important for the normal development of the child's brain. Maternal DHA supplementation increases DHA levels in mothers, but its direct link to infant neurodevelopment remains unclear. Obesity generates changes in the FA profile of pregnant women, causing an imbalance of n-3 and n-6 PUFA. An adequate level of DHA benefits children's cognitive function. However, a potential connection exists to the infants' inflammatory profile. CONCLUSION During gestation and lactation periods, an adequate DHA intake and n-6/ n-3 PUFA ratio (especially for obese women) are important for the optimal growth and brain development of the child.
Collapse
Affiliation(s)
- Yasna Muñoz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile; Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Chile
| | - Lorena Mercado
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile; Dirección de Postgrado, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370035, Chile
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Paz Beyer
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile; Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Chile
| | - Ignacio Alvear
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Francisca Echeverría
- Carrera Nutrición y Dietética, Departamento de Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
21
|
Shi P, Meng R, Xu J, Zhang Q, Ye G, Yan X, Liao K. Fatty acid translocase (FAT/CD36) in silver pomfret (Pampus argenteus): Molecular cloning and functional characterization. Comp Biochem Physiol B Biochem Mol Biol 2024; 270:110926. [PMID: 38036286 DOI: 10.1016/j.cbpb.2023.110926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Understanding the mechanisms of lipid transport and metabolism in fish is crucial to enhance dietary lipid utilization. Here, fatty acid translocase (CD36) gene was characterized in silver pomfret (Pampus argenteus). The open reading frame of silver pomfret cd36 gene was 1395 bp, encoding 464 amino acids. The silver pomfret CD36 protein contained typical transmembrane regions and N-glycosylation modification sites, and was localized to the cytomembrane. The cd36 gene was ubiquitously expressed in all tested tissues, with the highest expression observed in brain tissue. In vivo, both fasting and short-term high-fat feeding could increase cd36 expression in intestinal tissue. In vitro, cd36 expression was induced by palmitic acid, oleic acid, linolenic acid, eicosapentaenoic acid (EPA), and docosahexaenoic acid treatment in intestinal tissue. Furthermore, dual-luciferase reporter assay results indicated that peroxisome proliferator-activated receptor gamma (PPARγ) could enhance cd36 promoter activity, and the co-expression of cd36 and pparγ was observed in EPA-incubated intestine, suggesting that EPA may regulate the expression of cd36 via PPARγ to maintain the homeostasis of intestinal lipid metabolism in silver pomfret. These results highlighted the crucial role of CD36 in silver pomfret, and suggested that the cd36 expression may be regulated by PPARγ. This study could contribute to a greater understanding of lipid metabolism and the development of effective strategies for nutrient requirements in fish.
Collapse
Affiliation(s)
- Peng Shi
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Ran Meng
- Ningbo Academy of Oceanology and Fishery, Ningbo 315012, China
| | - Jilin Xu
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Qian Zhang
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Guochao Ye
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Xiaojun Yan
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Kai Liao
- School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
22
|
Kim OY, Song J. Important roles of linoleic acid and α-linolenic acid in regulating cognitive impairment and neuropsychiatric issues in metabolic-related dementia. Life Sci 2024; 337:122356. [PMID: 38123015 DOI: 10.1016/j.lfs.2023.122356] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Metabolic syndrome (MetS), which is characterized by insulin resistance, high blood glucose, obesity, and dyslipidemia, is known to increase the risk of dementia accompanied by memory loss and depression. The direct pathways and specific mechanisms in the central nervous system (CNS) for addressing fatty acid imbalances in MetS have not yet been fully elucidated. Among polyunsaturated acids, linoleic acid (LA, n6-PUFA) and α-linolenic acid (ALA, n3-PUFA), which are two essential fatty acids that should be provided by food sources (e.g., vegetable oils and seeds), have been reported to regulate various cellular mechanisms including apoptosis, inflammatory responses, mitochondrial biogenesis, and insulin signaling. Furthermore, inadequate intake of LA and ALA is reported to be involved in neuropathology and neuropsychiatric diseases as well as imbalanced metabolic conditions. Herein, we review the roles of LA and ALA on metabolic-related dementia focusing on insulin resistance, dyslipidemia, synaptic plasticity, cognitive function, and neuropsychiatric issues. This review suggests that LA and ALA are important fatty acids for concurrent treatment of both MetS and neurological problems.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Busan, Republic of Korea; Department of Health Sciences, Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Tao Z, Wang Y. The health benefits of dietary short-chain fatty acids in metabolic diseases. Crit Rev Food Sci Nutr 2024; 65:1579-1592. [PMID: 38189336 DOI: 10.1080/10408398.2023.2297811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Short-chain fatty acids (SCFAs) are a subset of fatty acids that play crucial roles in maintaining normal physiology and developing metabolic diseases, such as obesity, diabetes, cardiovascular disease, and liver disease. Even though dairy products and vegetable oils are the direct dietary sources of SCFAs, their quantities are highly restricted. SCFAs are produced indirectly through microbial fermentation of fibers. The biological roles of SCFAs in human health and metabolic diseases are mainly due to their receptors, GPR41 and GPR43, FFAR2 and FFAR3. Additionally, it has been demonstrated that SCFAs modulate DNMTs and HDAC activities, inhibit NF-κB-STAT signaling, and regulate G(i/o)βγ-PLC-PKC-PTEN signaling and PPARγ-UCP2-AMPK autophagic signaling, thus mitigating metabolic diseases. Recent studies have uncovered that SCFAs play crucial roles in epigenetic modifications of DNAs, RNAs, and post-translational modifications of proteins, which are critical regulators of metabolic health and diseases. At the same time, dietary recommendations for the purpose of SCFAs have been proposed. The objective of the review is to summarize the most recent research on the role of dietary SCFAs in metabolic diseases, especially the signal transduction of SCFAs in metabolic diseases and their functional efficacy in different backgrounds and models of metabolic diseases, at the same time, to provide dietary and nutritional recommendations for using SCFAs as food ingredients to prevent metabolic diseases.
Collapse
Affiliation(s)
- Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Nutrition Sciences, Texas Woman's University, Denton, Texas, USA
| | - Yao Wang
- Diabetes Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
24
|
Jayathilake AG, Luwor RB, Nurgali K, Su XQ. Molecular Mechanisms Associated with the Inhibitory Role of Long Chain n-3 PUFA in Colorectal Cancer. Integr Cancer Ther 2024; 23:15347354241243024. [PMID: 38708673 PMCID: PMC11072084 DOI: 10.1177/15347354241243024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/14/2024] [Accepted: 03/11/2024] [Indexed: 05/07/2024] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death in the world. Multiple evidence suggests that there is an association between excess fat consumption and the risk of CRC. The long chain n-3 polyunsaturated fatty acids (LC n-3 PUFA), especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are essential for human health, and both in vitro and in vivo studies have shown that these fatty acids can prevent CRC development through various molecular mechanisms. These include the modulation of arachidonic acid (AA) derived prostaglandin synthesis, alteration of growth signaling pathways, arrest of the cell cycle, induction of cell apoptosis, suppression of angiogenesis and modulation of inflammatory response. Human clinical studies found that LC n-3 PUFA combined with chemotherapeutic agents can improve the efficacy of treatment and reduce the dosage of chemotherapy and associated side effects. In this review, we discuss comprehensively the anti-cancer effects of LC n-3 PUFA on CRC, with a main focus on the underlying molecular mechanisms.
Collapse
Affiliation(s)
| | - Rodney Brain Luwor
- The University of Melbourne, Melbourne, VIC, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne, VIC, Australia
- Australian Institute for Muscular Skeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Xiao Qun Su
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Ortiz M, Álvarez D, Muñoz Y, Crisosto N, Valenzuela R, Maliqueo M. Linoleic and Arachidonic Fatty Acids and their Potential Relationship with Inflammation, Pregnancy, and Fetal Development. Curr Med Chem 2024; 31:5046-5060. [PMID: 37415369 DOI: 10.2174/0929867331666230706161144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 07/08/2023]
Abstract
A healthy maternal diet must consider an appropriate supply of long-chain polyunsaturated fatty acids (LCPUFAs) precursors to ensure adequate growth and development of the fetus. In this regard, n-6 PUFAs, predominantly linoleic (C18:2 n-6, LA) and arachidonic acid (C20:4 n-6), have a central role in the development of the central nervous system because they are part of the membrane structure and participate in the metabolism and signal transduction of cells. Nevertheless, they can also be transformed into inflammatory metabolites promoting the pathogenesis of cardiovascular diseases, cancer, and autoimmune or inflammatory conditions. In modern westernized societies, there is a high dietary consumption of foods rich in n-6 PUFAs which could have detrimental consequences for the fetus and neonate due to excessive exposure to these fatty acids (FAs). OBJECTIVE To summarize the evidence of maternal, placental, and fetal alterations that an excessive intake of n-6 polyunsaturated FAs (PUFAs), LA, and AA, could produce during pregnancy. METHODS A thorough review of the literature regarding the effects of n-6 PUFAs during pregnancy and lactation including in vivo and in vitro models, was carried out using the PubMed database from the National Library of Medicine-National Institutes of Health. RESULTS An elevated intake of n-6 PUFA, specifically LA, during pregnancy influences children's motor, cognitive, and verbal development during infancy and early childhood. Similarly, they could harm the placenta and the development of other fetal organs such as the fat tissue, liver, and cardiovascular system. CONCLUSION Maternal diet, specifically LA intake, could have significant repercussions on fetal development and long-term consequences in the offspring, including the possibility of future metabolic and mental diseases. It would be necessary to focus on the prevention of these alterations through timely dietary interventions in the target population.
Collapse
Affiliation(s)
- Macarena Ortiz
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago, Chile
| | - Daniela Álvarez
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago, Chile
| | - Yasna Muñoz
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago, Chile
| | - Nicolás Crisosto
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago, Chile
- Endocrinology Unit, Department of Medicine, Clínica Alemana de Santiago, Faculty of Medicine, Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Rodrigo Valenzuela
- Nutrition Department, School of Medicine, Universidad de Chile, Santiago, Chile
| | - Manuel Maliqueo
- Laboratory of Endocrinology and Metabolism, Department of Medicine West Division, Universidad de Chile, Santiago, Chile
| |
Collapse
|
26
|
Sztolsztener K, Bzdęga W, Konstantynowicz-Nowicka K, Chabowski A, Harasim-Symbor E. Therapeutic effect of cannabidiol on myocardial arachidonic acid content in various lipid fractions in a rat model of obesity. Prostaglandins Other Lipid Mediat 2023; 169:106767. [PMID: 37541613 DOI: 10.1016/j.prostaglandins.2023.106767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
The study explored the potential protective influence of cannabidiol (CBD) on myocardial inflammation state, with a special focus on arachidonic acid (AA), and oxidative balance in lipid overload conditions. The 7-week experiment was conducted on male Wistar rats receiving standard or high-fat diet (HFD) with intraperitoneal CBD injections for the last 14 days. The n-3 and n-6 polyunsaturated fatty acids (PUFAs) activities and AA concentration in selected fractions were evaluated by gas-liquid chromatography (GLC). The expression of proteins was determined by Western blot and the concentration of different parameters by ELISA, colorimetric, or multiplex assay kits. Our results revealed that CBD increased n-3 PUFAs activity in phospholipid and triacylglycerol fractions, and decreased AA content in the HFD group, especially in the phospholipid pool. Simultaneously, CBD decreased the expression of nuclear factor kappa B, cyclooxygenase-1, and - 2, resulting in the reduction of prostaglandin E2 and the increment of prostaglandin I2. CBD appears to be relatively safe for the treatment of obesity-induced heart disease, as it has anti-inflammatory and partially antioxidative properties.
Collapse
Affiliation(s)
- Klaudia Sztolsztener
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, 15-222 Bialystok, Poland
| | - Wiktor Bzdęga
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, 15-222 Bialystok, Poland
| | | | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, 15-222 Bialystok, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Mickiewicz Str. 2C, 15-222 Bialystok, Poland.
| |
Collapse
|
27
|
Luan M, Wang J, Liang K, Li B, Liu K. Association between the intake of dietary n3 and n6 fatty acids and stroke in US adults: A cross-sectional study of NHANES 2007-2018. PLoS One 2023; 18:e0293893. [PMID: 37971997 PMCID: PMC10653462 DOI: 10.1371/journal.pone.0293893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/21/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The association between the intake of dietary n3 and n6 fatty acids and the risk of stroke is subject to debate. The primary objective of the present research was to establish the correlation in a large sample of American adults. METHODS Using data from the National Health and Nutrition Examination Survey (NHANES) between 2007 and 2018, the association of the intake of dietary n3 and n6 fatty acids with stroke events was analyzed in a sample of 29,459 adults. The intake of n3 and n6 fatty acids intake was assessed though two 24-h dietary recalls. Stroke outcomes were identified based on the responses provided in self-reported questionnaire. Logistic regression was fitted to evaluate the correlation of dietary n3, n6 fatty acids intake with stroke events. RESULTS Subjects in the highest tertile (T3) of dietary n3 (OR: 0.67, 95% CI: 0.49-0.93), n6 (OR: 0.65, 95% CI: 0.45-0.95) fatty acids intake were found to have obviously lower risk of stroke compared to those in the lowest tertile (T1), but the n6:n3 ratio was not found to be associated with a stroke event. Results from stratified analysis demonstrated that dietary n3 fatty acids had an inverse correlation of stroke in both male and female, but dietary n6 fatty acids only had this correlation in male. Moreover, findings were made that the interaction was significant in terms of age in the subgroup analysis, and the negative relationship between the intake of dietary n3 and n6 fatty acids and stroke event were particularly pronounced among participants aged ≥60. CONCLUSIONS The present results suggested that increased dietary n3, n6 fatty acids intake correlated with a lower risk of stroke.
Collapse
Affiliation(s)
- Mingya Luan
- General Practice Department of Medicine, 960th Hospital People’s Liberation Army of China Joint Logist Support Force, Jinan, China
| | - Jia Wang
- Public Health Department, Weihai Maternal and Child Health Hospital, the Affiliated Weihai Second Municipal Hospital of Qingdao University, Weihai, China
| | - Kun Liang
- General Practice Department of Medicine, 960th Hospital People’s Liberation Army of China Joint Logist Support Force, Jinan, China
| | - Bo Li
- Zhangcun Town Health Center in Huancui District, Weihai, China
| | - Kewei Liu
- General Practice Department of Medicine, 960th Hospital People’s Liberation Army of China Joint Logist Support Force, Jinan, China
| |
Collapse
|
28
|
Meng X, Yu G, Luo T, Zhang R, Zhang J, Liu Y. Transcriptomics integrated with metabolomics reveals perfluorobutane sulfonate (PFBS) exposure effect during pregnancy and lactation on lipid metabolism in rat offspring. CHEMOSPHERE 2023; 341:140120. [PMID: 37696479 DOI: 10.1016/j.chemosphere.2023.140120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023]
Abstract
Emerging epidemiological evidence indicates potential associations between gestational perfluorobutane sulfonate (PFBS) exposure and adverse metabolic outcomes in offspring. However, the underlying mechanisms remain unclear. Our study aimed to investigate PFBS exposure effects during pregnancy and lactation on rat offspring lipid profiles and the possible underlying mechanisms. Although the biochemical index difference including total cholesterol (TC), triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), alanine amino transaminase (ALT), aspartate amino transferase (AST), and fasting blood glucose between exposed groups and the control group was not significant, transcriptome analyses showed that the differentially expressed genes (DEGs) in the 50 mg/kg/day PFBS exposure group were significantly related to protein digestion and absorption, peroxisome proliferator activated-receptor (PPAR) signaling pathway, xenobiotic metabolism by cytochrome P450, glycine, serine and threonine metabolism, β-alanine metabolism, bile secretion, unsaturated fatty acid (FA) biosynthesis, and alanine, aspartate and glutamate metabolism. Untargeted metabolomics analyses identified 17 differential metabolites in the 50 mg/kg/day PFBS exposure group. Among these, phosphatidylserine [PS (18:0/22:6(4Z,7Z,10Z,13Z,16Z,19Z))], lysoPE (18:1(11Z)/0:0), and PS (14:0/20:4(5Z,8Z,11Z,14Z)) were significantly correlated with phospholipid metabolism disorders. Correlation analysis indicated the DEGs, including FA binding protein (Fabp4), spermine oxidase (Smox), Fabp2, acyl-CoA thioesterase 5 (Acot5), sarcosine dehydrogenase (Sardh), and amine oxidase, copper-containing 3 (Aoc3) that significantly enriched in xenobiotic metabolism by cytochrome P450 and glycine, serine, and threonine metabolism signaling pathways were highly related to the differential metabolite pantetheine 4'-phosphate. Pantetheine 4'-phosphate was significantly negatively associated with non-high-density lipoprotein (non-HDL) and TC levels. Collectively, our study indicated that maternal PFBS exposure at a relatively low level could alter gene expression and metabolic molecules in lipid metabolism-related pathway series in rat offspring, although the effects on metabolic phenotypes were not significant within the limited observational period, using group-wise and trend analyses.
Collapse
Affiliation(s)
- Xi Meng
- Ministry of Education -Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guoqi Yu
- Ministry of Education -Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Global Center for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Tingyu Luo
- School of Public Health, Guilin Medical University, Guilin, 541001, China
| | - Ruiyuan Zhang
- Ministry of Education -Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jun Zhang
- Ministry of Education -Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yongjie Liu
- Ministry of Education -Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
29
|
Li X, Qi F, Zhao Z, Ma J. Association of dietary fatty acid intake with hypertension in children and adolescents: evidence from the NHANES 2005-2018. Front Pediatr 2023; 11:1185982. [PMID: 37753192 PMCID: PMC10518386 DOI: 10.3389/fped.2023.1185982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
Aim This study aims to evaluate the association between dietary fatty acid intake and hypertension in children and adolescents. Methods This cross-sectional study used data of children and adolescents aged 8-17 years from the National Health and Nutrition Examination Survey (NHANES) 2005-2018. Dietary intake of total fat and fatty acid was evaluated via two 24-h dietary recall interviews. Multivariate logistic regression models were used to assess the association between fatty acid intake and hypertension, with odds ratios (ORs) and 95% confidence intervals (CIs) calculated. A subgroup analysis was conducted according to gender, age, and body mass index Z-score. Results This study included 13,330 subjects, of which 11,614 were non-hypertensive and 1,716 were hypertensive. Higher intake of total polyunsaturated fatty acids (PUFAs) was associated with significantly lower odds of hypertension (OR = 0.85, 95% CI: 0.74-0.97, P = 0.018). No significant associations were found between the density of total saturated fatty acid, monounsaturated fatty acids, and PUFAs and the odds of hypertension (all P > 0.05). Increased intake of omega-3 (OR = 0.82, 95% CI: 0.72-0.93, P = 0.002) and omega-6 (OR = 0.86, 95% CI: 0.75-0.98, P = 0.025) PUFAs, octadecatrienoic acid (OR = 0.82, 95% CI: 0.72-0.93, P = 0.003), and octadecadienoic acid (OR = 0.86, 95% CI: 0.75-0.98, P = 0.025) was associated with significantly lower odds of hypertension, and individuals with higher omega-6/omega-3 ratio had significantly higher odds of hypertension (OR = 1.09, 95% CI: 1.02-1.17, P = 0.025). The density of omega-3 PUFAs (OR = 0.86, 95% CI: 0.78-0.95, P = 0.004) and octadecatrienoic acid (OR = 0.87, 95% CI: 0.78-0.96, P = 0.006) was inversely associated with the odds of hypertension, and the omega-6/omega-3 ratio was positively associated with the odds of hypertension (OR = 1.09, 95% CI: 1.02-1.17, P = 0.012). Conclusion Total PUFA intake was negatively associated with the odds of hypertension in children and adolescents. Higher intake of omega-3 and omega-6 PUFAs, octadecatrienoic acid, and octadecadienoic acid, as well as density of omega-3 PUFAs and octadecatrienoic acid, was associated with lower odds of hypertension.
Collapse
Affiliation(s)
- Xiumin Li
- Department of Pediatric Medicine, The Second People’s Hospital of Liaocheng, Linqing, China
| | - Fengqin Qi
- Department of Pediatric Medicine, The Second People’s Hospital of Liaocheng, Linqing, China
| | - Zhihong Zhao
- Department of Pediatric Medicine, The Second People’s Hospital of Liaocheng, Linqing, China
| | - Jinbang Ma
- Department of Neurosurgery, The Second People’s Hospital of Liaocheng, Linqing, China
| |
Collapse
|
30
|
Muthuswamy K, Shanmugamprema D, Subramanian G, Ponnusamy V, Vasanthakumar K, Krishnan V, Palanivelu PR, Rajasekaran S, Subramaniam S. CD36 genetic polymorphism and salivary cues are associated with oleic acid sensitivity and dietary fat intake. NUTR BULL 2023; 48:376-389. [PMID: 37533360 DOI: 10.1111/nbu.12633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/07/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
There is a lack of research on the combined effects of genetic variations (specifically CD36 SNPs-rs1761667 and rs1527483), dietary food habits (vegetarian or not), and the salivary environment on obesity and taste sensitivity, especially in the Indian population. The current study aims to better understand the relationship between impaired taste perception, fat consumption, higher BMI and obesity development by examining the combined association between CD36 SNPs, oleic acid (OA) detection threshold, and food habits among Indian participants. Furthermore, the relationship between oral fatty acid (FAs) sensitivity and taste physiology factors linked to inflammation and salivary proteins was considered. Participants with the minor allele (AA/AG) of CD36 (in both rs1527483 and rs1761667) consumed more fat, particularly saturated FAs (p = 0.0351). Salivary lipopolysaccharide, which causes inflammation, was significantly greater in non-vegetarians with a higher BMI (p < 0.05), and it exhibited a negative correlation (r = -0.232 and p < 0.05) with Ki67 gene expression, a marker for taste progenitor cells. A positive correlation (r = 0.474, p = 0.04) between TLR4 mRNA levels and the OA detection threshold was also observed. Participants with BMI > 25 kg/m2 had substantially higher TNF-α and IL-6 receptor mRNA expression levels, but there were no significant differences between the vegetarian and non-vegetarian groups. However, salivary CA-VI, which has a buffering capability on the oral environment, was lower in non-vegetarian adults with BMI >25. Thus, it was shown that non-vegetarians with overweight and obesity in India were in at-risk groups for the CD36 SNP (AA/AG at rs1761667 and rs1527483) and had higher levels of inflammatory markers, which exacerbated alterations in food behaviour and physiological changes, indicating their relevance in the development of obesity.
Collapse
Affiliation(s)
- Karthi Muthuswamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, India
| | | | - Gowtham Subramanian
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, India
| | - Vinithra Ponnusamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, India
| | - Keerthana Vasanthakumar
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, India
| | - Vasanth Krishnan
- Molecular Biology Laboratory, Department of Botany, Bharathiar University, Coimbatore, India
| | | | | | - Selvakumar Subramaniam
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, India
| |
Collapse
|
31
|
Sun J, Bian Y, Ma Y, Ali W, Wang T, Yuan Y, Gu J, Bian J, Liu Z, Zou H. Melatonin alleviates cadmium-induced nonalcoholic fatty liver disease in ducks by alleviating autophagic flow arrest via PPAR-α and reducing oxidative stress. Poult Sci 2023; 102:102835. [PMID: 37343350 PMCID: PMC10404762 DOI: 10.1016/j.psj.2023.102835] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
Cadmium (Cd) is an important environmental pollutant that causes liver damage and induces nonalcoholic fatty liver disease (NAFLD). NAFLD is a fat accumulation disease and has significant effects on the body. Melatonin (Mel) is an endogenous protective molecule with antioxidant, anti-inflammatory, antiobesity, and antiaging effects. However, whether Mel can alleviate Cd-induced NAFLD and its mechanism remains unclear. First, in vivo, we found that Mel maintained mitochondrial structure and function, inhibited oxidative stress, and reduced Cd-induced liver injury. In addition, Mel alleviated lipid accumulation in the liver induced by Cd. In this process, Mel inhibits fatty acid production and promotes fatty acid oxidation. Interestingly, Mel regulated PPAR-α expression and alleviated Cd-induced autophagy blockade. In vitro model, the oil Red O staining, and WB results showed that Mel alleviated Cd-induced lipid accumulation. In addition, RAPA was used to activate autophagy to alleviate Cd-induced lipid accumulation, and TG was used to block autophagy flux to aggravate Cd-induced autophagy accumulation. After knocking down PPAR-α, the autophagosome fusion with lysosomes, and autophagic flux was inhibited and increased Cd-induced lipid accumulation. Mel alleviates mitochondrial damage and oxidative stress, and attenuates Cd-induced NAFLD by restoring the expression of PPAR-α and restoring autophagy flux.
Collapse
Affiliation(s)
- Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yusheng Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Waseem Ali
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
32
|
Phung NV, Rong F, Xia WY, Fan Y, Li XY, Wang SA, Li FL. Nervonic acid and its sphingolipids: Biological functions and potential food applications. Crit Rev Food Sci Nutr 2023; 64:8766-8785. [PMID: 37114919 DOI: 10.1080/10408398.2023.2203753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Nervonic acid, a 24-carbon fatty acid with only one double bond at the 9th carbon (C24:1n-9), is abundant in the human brain, liver, and kidney. It not only functions in free form but also serves as a critical component of sphingolipids which participate in many biological processes such as cell membrane formation, apoptosis, and neurotransmission. Recent studies show that nervonic acid supplementation is not only beneficial to human health but also can improve the many medical conditions such as neurological diseases, cancers, diabetes, obesity, and their complications. Nervonic acid and its sphingomyelins serve as a special material for myelination in infants and remyelination patients with multiple sclerosis. Besides, the administration of nervonic acid is reported to reduce motor disorder in mice with Parkinson's disease and limit weight gain. Perturbations of nervonic acid and its sphingolipids might lead to the pathogenesis of many diseases and understanding these mechanisms is critical for investigating potential therapeutic approaches for such diseases. However, available studies about this aspect are limited. In this review, relevant findings about functional mechanisms of nervonic acid have been comprehensively and systematically described, focusing on four interconnected functions: cellular structure, signaling, anti-inflammation, lipid mobilization, and their related diseases.
Collapse
Affiliation(s)
- Nghi Van Phung
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Fei Rong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wan Yue Xia
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Yong Fan
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Xian Yu Li
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Shi An Wang
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Shandong Energy Institute, Qingdao, China
| | - Fu Li Li
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
- Shandong Energy Institute, Qingdao, China
| |
Collapse
|
33
|
Grajchen E, Loix M, Baeten P, Côrte-Real BF, Hamad I, Vanherle S, Haidar M, Dehairs J, Broos JY, Ntambi JM, Zimmermann R, Breinbauer R, Stinissen P, Hellings N, Verberk SGS, Kooij G, Giera M, Swinnen JV, Broux B, Kleinewietfeld M, Hendriks JJA, Bogie JFJ. Fatty acid desaturation by stearoyl-CoA desaturase-1 controls regulatory T cell differentiation and autoimmunity. Cell Mol Immunol 2023; 20:666-679. [PMID: 37041314 DOI: 10.1038/s41423-023-01011-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/23/2023] [Indexed: 04/13/2023] Open
Abstract
The imbalance between pathogenic and protective T cell subsets is a cardinal feature of autoimmune disorders such as multiple sclerosis (MS). Emerging evidence indicates that endogenous and dietary-induced changes in fatty acid metabolism have a major impact on both T cell fate and autoimmunity. To date, however, the molecular mechanisms that underlie the impact of fatty acid metabolism on T cell physiology and autoimmunity remain poorly understood. Here, we report that stearoyl-CoA desaturase-1 (SCD1), an enzyme essential for the desaturation of fatty acids and highly regulated by dietary factors, acts as an endogenous brake on regulatory T-cell (Treg) differentiation and augments autoimmunity in an animal model of MS in a T cell-dependent manner. Guided by RNA sequencing and lipidomics analysis, we found that the absence of Scd1 in T cells promotes the hydrolysis of triglycerides and phosphatidylcholine through adipose triglyceride lipase (ATGL). ATGL-dependent release of docosahexaenoic acid enhanced Treg differentiation by activating the nuclear receptor peroxisome proliferator-activated receptor gamma. Our findings identify fatty acid desaturation by SCD1 as an essential determinant of Treg differentiation and autoimmunity, with potentially broad implications for the development of novel therapeutic strategies and dietary interventions for autoimmune disorders such as MS.
Collapse
Affiliation(s)
- Elien Grajchen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Paulien Baeten
- University MS Center Hasselt, Pelt, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Beatriz F Côrte-Real
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium
| | - Ibrahim Hamad
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium
| | - Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Mansour Haidar
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI - Leuven Cancer Institute, KU Leuven - University of Leuven, Leuven, Belgium
| | - Jelle Y Broos
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam, The Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - James M Ntambi
- Department of Biochemistry, Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, USA
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Rolf Breinbauer
- BioTechMed-Graz, Graz, Austria
- Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Piet Stinissen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Niels Hellings
- University MS Center Hasselt, Pelt, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sanne G S Verberk
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI - Leuven Cancer Institute, KU Leuven - University of Leuven, Leuven, Belgium
| | - Bieke Broux
- University MS Center Hasselt, Pelt, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- Cardiovascular Research Institute Maastricht, Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Markus Kleinewietfeld
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
- University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
34
|
Bakker N, Hickey M, Shams R, Rivera CF, Vlahos J, Cense HA, Demirkiran A, Ramkhelawon B, Houdijk AP. Oral ω-3 PUFA supplementation modulates inflammation in adipose tissue depots in morbidly obese women: A randomized trial. Nutrition 2023; 111:112055. [PMID: 37182400 DOI: 10.1016/j.nut.2023.112055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/16/2023] [Accepted: 04/04/2023] [Indexed: 05/16/2023]
Abstract
OBJECTIVES Obesity is characterized by local and systemic low-grade inflammatory responses. Adipose tissue macrophages (ATM) play decisive roles in inflammation, insulin signaling, and various metabolic dysfunctions. Diets enriched with ω-3 polyunsaturated fatty acids (PUFAs) have been shown to improve health and mitigate pathologic conditions. However, the effects of ω-3 PUFA on adipose tissue inflammation, ATM number, and phenotype are poorly defined in human obesity. The aim of this study was to examine differences in expression of metabolic-inflammatory markers in omental, mesenteric, and subcutaneous fat depots of obese women supplemented with ω-3 PUFAs for 4 wk compared with a low-calorie diet before bariatric surgery. METHODS In a randomized controlled trial, inflammatory markers in the abdominal adipose tissue and the systemic response in obese women were studied. Patients were treated with a 2-wk low-calorie diet (LCD) or a 4-wk ω-3 PUFA-enriched diet (920 mg eicosapentaenoic acid, 760 mg docosahexaenoic acid daily) before laparoscopic bypass surgery. Omental, mesenteric, and subcutaneous adipose tissue biopsies were collected during surgery and analyzed for quantity and phenotype of ATMs, and profiled for adipokines, cytokines, and signal transduction molecules. RESULTS The chronic inflammatory state characterized by ATM markers was mostly improved by ω-3 PUFAs in visceral adipose tissue. We observed a decreased expression of CD45, CCL2, and CD68, indicating a lower inflammatory state. In patients with type 2 diabetes, ω-3 PUFAs lowered the expression of Netrin-1. CONCLUSIONS Compared with an LCD, a diet enriched with ω-3 PUFAs influences the inflammatory state in different adipose tissue depots, by affecting markers of adipose tissue inflammation, macrophage phenotype, and retention. However, this was not reflected in clinical parameters such as insulin resistance and inflammatory cytokines. Subcutaneous adipose tissue and visceral adipose tissue have different responses to an LCD or a ω-3 PUFA-enriched diet. The presence of diabetes modifies the expression of inflammatory markers.
Collapse
Affiliation(s)
- Nathalie Bakker
- Northwest Clinics, Department of Surgery, Alkmaar, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Red Cross Hospital, Department of Surgery, Beverwijk, The Netherlands
| | - Meave Hickey
- Division of Vascular and Endovascular Surgery, Department of Surgery and Cell Biology, New York University Langone Medical Center, New York, New York, United States
| | - Rebecca Shams
- Division of Vascular and Endovascular Surgery, Department of Surgery and Cell Biology, New York University Langone Medical Center, New York, New York, United States
| | - Cristobal F Rivera
- Division of Vascular and Endovascular Surgery, Department of Surgery and Cell Biology, New York University Langone Medical Center, New York, New York, United States
| | - John Vlahos
- Division of Vascular and Endovascular Surgery, Department of Surgery and Cell Biology, New York University Langone Medical Center, New York, New York, United States
| | - Huib A Cense
- Red Cross Hospital, Department of Surgery, Beverwijk, The Netherlands
| | - Ahmet Demirkiran
- Red Cross Hospital, Department of Surgery, Beverwijk, The Netherlands
| | - Bhama Ramkhelawon
- Division of Vascular and Endovascular Surgery, Department of Surgery and Cell Biology, New York University Langone Medical Center, New York, New York, United States
| | - Alexander Pj Houdijk
- Northwest Clinics, Department of Surgery, Alkmaar, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Amsterdam, The Netherlands; Red Cross Hospital, Department of Surgery, Beverwijk, The Netherlands.
| |
Collapse
|
35
|
Wei XF, Liu YJ, Li SW, Ding L, Han SC, Chen ZX, Lu H, Wang P, Sun YC. Stress response and tolerance mechanisms of NaHCO 3 exposure based on biochemical assays and multi-omics approach in the liver of crucian carp (Carassius auratus). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114633. [PMID: 36889228 DOI: 10.1016/j.ecoenv.2023.114633] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
The development and utilization of saline-alkaline water, an important backup resource, has received widespread attention. However, the underuse of saline-alkaline water, threatened by the single species of saline-alkaline aquaculture, seriously affects the development of the fishery economy. In this work, a 30-day NaHCO3 stress experimental study combined with analyses of untargeted metabolomics, transcriptome, and biochemical approaches was conducted on crucian carp to provide a better understanding of the saline-alkaline stress response mechanism in freshwater fish. This work revealed the relationships among the biochemical parameters, endogenous differentially expressed metabolites (DEMs), and differentially expressed genes (DEGs) in the crucian carp livers. The biochemical analysis showed that NaHCO3 exposure changed the levels of several physiological parameters associated with the liver, including antioxidant enzymes (SOD, CAT, GSH-Px), MDA, AKP, and CPS. According to the metabolomics study, 90 DEMs are involved in various metabolic pathways such as ketone synthesis and degradation metabolism, glycerophospholipid metabolism, arachidonic acid metabolism, and linoleic acid metabolism. In addition, transcriptomics data analysis showed that a total of 301 DEGs were screened between the control group and the high NaHCO3 concentration group, of which 129 up-regulated genes and 172 down-regulated genes. Overall, NaHCO3 exposure could cause lipid metabolism disorders and induce energy metabolism imbalance in the crucian carp liver. Simultaneously, crucian carp might regulate its saline-alkaline resistance mechanism by enhancing the synthesis of glycerophospholipid metabolism, ketone bodies, and degradation metabolism, at the same time increasing the vitality of antioxidant enzymes (SOD, CAT, GSH-Px) and nonspecific immune enzyme (AKP). Herein, all results will provide new insights into the molecular mechanisms underlying the stress responses and tolerance to saline-alkaline exposure in crucian carp.
Collapse
Affiliation(s)
- Xiao-Feng Wei
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Ying-Jie Liu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shan-Wei Li
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Lu Ding
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shi-Cheng Han
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Zhong-Xiang Chen
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China
| | - Hang Lu
- College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Peng Wang
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China.
| | - Yan-Chun Sun
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; College of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
36
|
Yang JY, Zhang L, Zhang TT, Wang CC, Zhao YC, Li XY, Wang YM, Xue CH. Eicosapentaenoic acid-enriched phospholipids alleviate glucose and lipid metabolism in spontaneously hypertensive rats with CD36 mutation: a precise nutrition strategy. Food Funct 2023; 14:2349-2361. [PMID: 36843452 DOI: 10.1039/d2fo03016k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Previous studies have found that eicosapentaenoic acid-enriched phospholipids (EPA-PLs) alleviated glucose and lipid metabolism, which was accompanied by an increase of cluster of differentiation 36 (CD36). However, the effects of EPA-PLs on glucose and lipid metabolism in the case of CD36 mutation are unclear. Thus, spontaneously hypertensive rats/NCrl (SHR) were used as a CD36 mutation model to determine the effects of dietary 2% EPA-PLs for 4 weeks on glucose and lipid metabolism. The results showed that the intervention of EPA-PLs significantly alleviated the abnormal increase of serum free fatty acid levels and glycerol levels in SHRs. Moreover, the administration of EPA-PLs decreased the triglyceride levels and cholesterol levels by 31.1% and 37.9%, respectively, in the liver. Dietary EPA-PLs had no effect on epididymal fat weight, but EPA-PLs inhibited adipocyte hypertrophy in SHRs. Further mechanistic research found that EPA-PL pretreatment significantly reduced triacylglycerol catabolism and increased fatty acid β-oxidation. Additionally, the administration of EPA-PLs decreased the area under the curve of the intraperitoneal glucose tolerance test and fasting serum insulin levels by activating the IRS/PI3K/AKT signaling pathway. Furthermore, EPA-PL pretreatment significantly increased the CD36 gene expression in the liver tissues, adipose tissues and muscle tissues even in the case of CD36 mutation. These results indicated that EPA-PLs alleviate glucose and lipid metabolism in the case of CD36 mutation, which provides a precise nutrition strategy for people with CD36 mutation.
Collapse
Affiliation(s)
- Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China.
| | - Lingyu Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China. .,College of Food and Biological Engineering, Jimei University, Xiamen, 361021, Fujian, People's Republic of China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China.
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China.
| | - Ying-Cai Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China.
| | - Xiao-Yue Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China.
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China. .,Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, Shandong Province, People's Republic of China.
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, Shandong, People's Republic of China. .,Laboratory of Marine Drugs & Biological Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, Shandong Province, People's Republic of China.
| |
Collapse
|
37
|
Rodway LA, Pauls SD, Pascoe CD, Aukema HM, Taylor CG, Zahradka P. Distinct effects of α-linolenic acid and docosahexaenoic acid on the expression of genes related to cholesterol metabolism and the response to infection in THP-1 monocytes and immune cells of obese humans. Biomed Pharmacother 2023; 159:114167. [PMID: 36621145 DOI: 10.1016/j.biopha.2022.114167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Monocytes play a large role in chronic inflammatory conditions such as obesity, atherosclerosis and infection. Marine-derived omega-3 fatty acids such as docosahexaenoic acid (DHA) beneficially alter immune function and attenuate chronic inflammation in part by modifying gene expression. Comparisons with plant-derived omega-3 α-linolenic acid (ALA) on immune cell gene expression and function are limited. METHODS Transcriptome analysis was performed on THP-1 human monocytes treated with ALA, DHA or vehicle for 48 hr using fold change analysis, principal component analysis (PCA), partial least squares-discriminant analysis (PLS-DA), variable importance analysis (VIP), and ingenuity pathway analysis (IPA). Candidate genes were validated by qPCR. Functional assays evaluated the transcriptomic predictions. Expression of candidate transcripts identified in THP-1 cells were examined in PBMC from clinical trial (OXBIO; NCT03583281) participants consuming ALA- or DHA-rich oil supplements. FINDINGS ALA and DHA-treated monocytes presented distinct transcriptomic profiles as per VIP and PLS-DA. Both fatty acids were predicted to reduce cellular cholesterol content, while ALA would uniquely increase response to infection and chemotactic signals. Functional assays revealed ALA and DHA decreased cholesterol content. DHA significantly decreased the response to infection and chemotaxis, but ALA had no effect. Candidate transcripts responded similarly in PBMC from n-3 PUFA supplemented women with obesity. CONCLUSION ALA and DHA differentially alter the transcription profiles and functions associated with the response to infection, chemotaxis, and cholesterol metabolism in mononuclear immune cells. Thus, they may uniquely affect related disease processes contributing to obesity, atherosclerosis, and the response to infection.
Collapse
Affiliation(s)
- Lisa A Rodway
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Samantha D Pauls
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Christopher D Pascoe
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Harold M Aukema
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Carla G Taylor
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Peter Zahradka
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, Faculty of Agricultural and Food Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| |
Collapse
|
38
|
Miranda CS, Silva-Veiga FM, Fernandes-da-Silva A, Guimarães Pereira VR, Martins BC, Daleprane JB, Martins FF, Souza-Mello V. Peroxisome proliferator-activated receptors-alpha and gamma synergism modulate the gut-adipose tissue axis and mitigate obesity. Mol Cell Endocrinol 2023; 562:111839. [PMID: 36581062 DOI: 10.1016/j.mce.2022.111839] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
AIM To evaluate the effects of single PPARα or PPARγ activation, and their synergism (combined PPARα/γ activation) upon the gut-adipose tissue axis, focusing on the endotoxemia and upstream interscapular brown adipose tissue (iBAT) function in high-saturated fat-fed mice. METHODS Male C57BL/6 mice received a control diet (C, 10% lipids) or a high-fat diet (HF, 50% lipids) for 12 weeks. Then, the HF group was divided to receive the treatments for four weeks: HFγ (pioglitazone, 10 mg/kg), HFα (WY-14643, 3.5 mg/kg), and HFα/γ (tesaglitazar, 4 mg/kg). RESULTS The HF group exhibited overweight, oral glucose intolerance, gut dysbiosis, altered gut permeability, and endotoxemia, culminating in iBAT whitening. The downregulation of LPS-Tlr4 signaling underpinned reduced inflammation and improved lipid metabolism in iBAT in the HFα/γ group, the unique to show normalized body mass and increased energy expenditure. CONCLUSION PPARα/γ synergism treated obesity by ameliorating the gut-adipose tissue axis, where restored gut microbiota and permeability controlled endotoxemia and rescued iBAT whitening through favored thermogenesis.
Collapse
Affiliation(s)
- Carolline Santos Miranda
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Flávia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Aline Fernandes-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vitória Regina Guimarães Pereira
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Bruna Cadete Martins
- Laboratory for Studies of Interactions Between Nutrition and Genetics (LEING), Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics (LEING), Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
39
|
Varma S, Molangiri A, Kona SR, Ibrahim A, Duttaroy AK, Basak S. Fetal Exposure to Endocrine Disrupting-Bisphenol A (BPA) Alters Testicular Fatty Acid Metabolism in the Adult Offspring: Relevance to Sperm Maturation and Quality. Int J Mol Sci 2023; 24:ijms24043769. [PMID: 36835180 PMCID: PMC9958878 DOI: 10.3390/ijms24043769] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Daily exposure to bisphenols can affect reproductive functions due to their pseudo-estrogenic and/or anti-androgenic effects. Testicular lipids contain high levels of polyunsaturated fatty acids necessary for sperm maturity, motility, and spermatogenesis. Whether prenatal exposure to bisphenols alters testicular fatty acid metabolism in adult offspring is unknown. Pregnant Wistar rats were gavaged from gestational day 4 to 21 with BPA and BPS (0.0, 0.4, 4.0, 40.0 μg/kg bw/day). Despite increased body and testis weight, the total testicular cholesterol, triglyceride, and plasma fatty acids were unaffected in the offspring. Lipogenesis was upregulated by increased SCD-1, SCD-2, and expression of lipid storage (ADRP) and trafficking protein (FABP4). The arachidonic acid, 20:4 n-6 (ARA) and docosapentaenoic acid, 22:5 n-6 (DPA) levels were decreased in the BPA-exposed testis, while BPS exposure had no effects. The expression of PPARα, PPARγ proteins, and CATSPER2 mRNA were decreased, which are important for energy dissipation and the motility of the sperm in the testis. The endogenous conversion of linoleic acid,18:2 n-6 (LA), to ARA was impaired by a reduced ARA/LA ratio and decreased FADS1 expression in BPA-exposed testis. Collectively, fetal BPA exposure affected endogenous long-chain fatty acid metabolism and steroidogenesis in the adult testis, which might dysregulate sperm maturation and quality.
Collapse
Affiliation(s)
- Saikanth Varma
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India
| | - Archana Molangiri
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India
| | - Suryam Reddy Kona
- Lipid Chemistry Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India
| | - Ahamed Ibrahim
- Lipid Chemistry Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0316 Oslo, Norway
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India
- Correspondence: ; Tel./Fax: +91-40-27197336
| |
Collapse
|
40
|
Saika A, Tiwari P, Nagatake T, Node E, Hosomi K, Honda T, Kabashima K, Kunisawa J. Mead acid inhibits retinol-induced irritant contact dermatitis via peroxisome proliferator-activated receptor alpha. Front Mol Biosci 2023; 10:1097955. [PMID: 36825199 PMCID: PMC9941550 DOI: 10.3389/fmolb.2023.1097955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Retinol is widely used in topical skincare products to ameliorate skin aging and treat acne and wrinkles; however, retinol and its derivatives occasionally have adverse side effects, including the induction of irritant contact dermatitis. Previously, we reported that mead acid (5,8,11-eicosatrienoic acid), an oleic acid metabolite, ameliorated skin inflammation in dinitrofluorobenzene-induced allergic contact hypersensitivity by inhibiting neutrophil infiltration and leukotriene B4 production by neutrophils. Here, we showed that mead acid also suppresses retinol-induced irritant contact dermatitis. In a murine model, we revealed that mead acid inhibited keratinocyte abnormalities such as keratinocyte hyperproliferation. Consistently, mead acid inhibited p38 MAPK (mitogen-activated protein kinase) phosphorylation, which is an essential signaling pathway in the keratinocyte hyperplasia induced by retinol. These inhibitory effects of mead acid were associated with the prevention of both keratinocyte hyperproliferation and the gene expression of neutrophil chemoattractants, including Cxcl1 and Cxcl2, and they were mediated by a PPAR (peroxisome proliferator-activated receptor)-α pathway. Our findings identified the anti-inflammatory effects of mead acid, the use of which can be expected to minimize the risk of adverse side effects associated with topical retinoid application.
Collapse
Affiliation(s)
- Azusa Saika
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Prabha Tiwari
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Eri Node
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Tetsuya Honda
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, Collaborative Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan,International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo, Japan,Graduate School of Medicine, Graduate School of Dentistry, Graduate School of Pharmaceutical Sciences, Graduate School of Science, Osaka University, Suita, Osaka, Japan,Department of Microbiology and Immunology, Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan,Research Organization for Nano and Life Innovation, Waseda University, Shinjuku, Tokyo, Japan,Graduate School of Biomedical and Health Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan,*Correspondence: Jun Kunisawa,
| |
Collapse
|
41
|
Tian A, Sun Z, Zhang M, Li J, Pan X, Chen P. Associations between dietary fatty acid patterns and non-alcoholic fatty liver disease in typical dietary population: A UK biobank study. Front Nutr 2023; 10:1117626. [PMID: 36824175 PMCID: PMC9942598 DOI: 10.3389/fnut.2023.1117626] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/19/2023] [Indexed: 02/10/2023] Open
Abstract
Background and Aims Dietary fatty acid composition is associated with non-alcoholic fatty liver disease (NAFLD). Few evidence had identified a clear role of dietary fatty acid composition of typical diet in NAFLD. We aimed to investigate the relationship between dietary patterns and NAFLD in populations with typical diets and to explore the effect of fatty acid composition in dietary patterns on NAFLD. Methods Principal component analysis was used to identify 4 dietary patterns in UK Biobank participants. Logistic regression was used to estimate the association between dietary patterns and NAFLD. Mediation analysis was performed to evaluate the extent to which the relationship between dietary patterns and NAFLD was explained by dietary fatty acid combinations, as surrogated by serum fatty acids measured by nuclear magnetic resonance. Results A dietary fatty acid pattern (DFP1) characterized by "PUFA enriched vegetarian" was negatively associated with NAFLD risk. Serum fatty acids were significantly associated with DFP1 and NAFLD. Mediation analysis showed SFA (27.8%, p < 0.001), PUFA (25.1%, p < 0.001), ω-6 PUFA (14.3%, p < 0.001), LA (15.6%, p < 0.001) and DHA (10%, p < 0.001) had a significant indirect effect on the association between DFP1 and NAFLD. A dietary pattern characterized by "PUFA enriched carnivore" (DFP2) was not associated with NAFLD risk. Conclusion A "PUFA enriched vegetarian" dietary pattern with increased LA and DHA, may be beneficial for the treatment or prevention of NAFLD, while a "PUFA enriched carnivore" dietary pattern may not be harmful to NAFLD.
Collapse
Affiliation(s)
- Aowen Tian
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China,Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Zewen Sun
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Miaoran Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China,Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Jiuling Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China,Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xingchen Pan
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Peng Chen
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China,Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,*Correspondence: Peng Chen, ✉
| |
Collapse
|
42
|
Waterpipe smoke inhalation potentiates cardiac oxidative stress, inflammation, mitochondrial dysfunction, apoptosis and autophagy in experimental hypertension. Biomed Pharmacother 2023; 158:114144. [PMID: 36916396 DOI: 10.1016/j.biopha.2022.114144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/04/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Cigarette smoking worsens the health of hypertensive patients. However, less is known about the actions and underlying mechanisms of waterpipe smoke (WPS) in hypertension. Therefore, we evaluated the effects of WPS inhalation in mice made hypertensive (HT) by infusing angiotensin II for six weeks. On day 14 of the infusion of angiotensin II or vehicle (normotensive; NT), mice were exposed either to air or WPS for four consecutive weeks. Each session was 30 min/day and 5 days/week. In NT mice, WPS increased systolic blood pressure (SBP) compared with NT air-exposed group. SBP increase was elevated in HT+WPS group versus either HT+air or NT+WPS. Similarly, the plasma levels of brain natriuretic peptide, C-reactive protein, 8-isoprostane and superoxide dismutase were increased in HT+WPS compared with either HT+air or NT+WPS. In the heart tissue, several markers of oxidative stress and inflammation were increased in HT+WPS group vs the controls. Furthermore, mitochondrial dysfunction in HT+WPS group was more affected than in the HT+air or HT+WPS groups. WPS inhalation in HT mice significantly increased cardiac DNA damage, cleaved caspase 3, expression of the autophagy proteins beclin 1 and microtubule-associated protein light chain 3B, and phosphorylated nuclear factor κ B, compared with the controls. Compared with HT+air mice, heart histology of WPS-exposed HT mice showed increased cardiomyocyte damage, neutrophilic and lymphocytic infiltration and focal fibrosis. We conclude that, in HT mice, WPS inhalation worsened hypertension, cardiac oxidative stress, inflammation, mitochondrial dysfunction, DNA damage, apoptosis and autophagy. The latter effects were associated with a mechanism involving NF-κB activation.
Collapse
|
43
|
Vitamin A: A Key Inhibitor of Adipocyte Differentiation. PPAR Res 2023; 2023:7405954. [PMID: 36776154 PMCID: PMC9908342 DOI: 10.1155/2023/7405954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/04/2023] Open
Abstract
Inhibiting adipocyte differentiation, the conversion of preadipocytes to mature functional adipocytes, might represent a new approach to treating obesity and related metabolic disorders. Peroxisome proliferator-activated receptor γ and CCAAT-enhancer-binding protein α are two master coregulators controlling adipogenesis both in culture and in vivo. Many recent studies have confirmed the relationship between retinoic acid (RA) and the conversion of embryonic stem cells into adipocytes; however, these studies have shown that RA potently blocks the differentiation of preadipocytes into mature adipocytes. Nevertheless, the functional role of RA in early tissue development and stem cell differentiation, including in adipose tissue, remains unclear. This study highlights transcription factors that block adipocyte differentiation and maintain preadipocyte status, focusing on those controlled by RA. However, some of these novel adipogenesis inhibitors have not been validated in vivo, and their mechanisms of action require further clarification.
Collapse
|
44
|
Danielli M, Perne L, Jarc Jovičić E, Petan T. Lipid droplets and polyunsaturated fatty acid trafficking: Balancing life and death. Front Cell Dev Biol 2023; 11:1104725. [PMID: 36776554 PMCID: PMC9911892 DOI: 10.3389/fcell.2023.1104725] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Lipid droplets are fat storage organelles ubiquitously distributed across the eukaryotic kingdom. They have a central role in regulating lipid metabolism and undergo a dynamic turnover of biogenesis and breakdown to meet cellular requirements for fatty acids, including polyunsaturated fatty acids. Polyunsaturated fatty acids esterified in membrane phospholipids define membrane fluidity and can be released by the activity of phospholipases A2 to act as ligands for nuclear receptors or to be metabolized into a wide spectrum of lipid signaling mediators. Polyunsaturated fatty acids in membrane phospholipids are also highly susceptible to lipid peroxidation, which if left uncontrolled leads to ferroptotic cell death. On the one hand, lipid droplets act as antioxidant organelles that control polyunsaturated fatty acid storage in triglycerides in order to reduce membrane lipid peroxidation, preserve organelle function and prevent cell death, including ferroptosis. On the other hand, lipid droplet breakdown fine-tunes the delivery of polyunsaturated fatty acids into metabolic and signaling pathways, but unrestricted lipid droplet breakdown may also lead to the release of lethal levels of polyunsaturated fatty acids. Precise regulation of lipid droplet turnover is thus essential for polyunsaturated fatty acid distribution and cellular homeostasis. In this review, we focus on emerging aspects of lipid droplet-mediated regulation of polyunsaturated fatty acid trafficking, including the management of membrane lipid peroxidation, ferroptosis and lipid mediator signaling.
Collapse
Affiliation(s)
| | | | | | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
45
|
Boyer BB, Wiener HW, Hopkins SE, Purnell JQ, O'Brien DM, Aliwarga T, Pomeroy JJ, Aslan JE, Thummel KE, Tiwari HK. Obesity-Associated Dyslipidemia Is Moderated by Habitual Intake of Marine-Derived n-3 Polyunsaturated Fatty Acids in Yup'ik Alaska Native People: A Cross-Sectional Mediation-Moderation Analysis. J Nutr 2023; 153:279-292. [PMID: 36913463 PMCID: PMC10196570 DOI: 10.1016/j.tjnut.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Obesity leads to insulin resistance, altered lipoprotein metabolism, dyslipidemia, and cardiovascular disease. The relationship between long-term intake of n-3 polyunsaturated fatty acids (n-3 PUFAs) and prevention of cardiometabolic disease remains unresolved. OBJECTIVES The aim of this study was to explore direct and indirect pathways between adiposity and dyslipidemia, and the degree to which n-3 PUFAs moderate adiposity-induced dyslipidemia in a population with highly variable n-3 PUFA intake from marine foods. METHODS In total, 571 Yup'ik Alaska Native adults (18-87 y) were enrolled in this cross-sectional study. The red blood cell (RBC) nitrogen isotope ratio (15N/14N, or NIR) was used as a validated objective measure of n-3 PUFA intake. EPA and DHA were measured in RBCs. Insulin sensitivity and resistance were estimated by the HOMA2 method. Mediation analysis was conducted to evaluate the contribution of the indirect causal path between adiposity and dyslipidemia mediated through insulin resistance. Moderation analysis was used to assess the influence of dietary n-3 PUFAs on the direct and indirect paths between adiposity and dyslipidemia. Outcomes of primary interest included plasma total cholesterol (TC), LDL-cholesterol (LDL-C), HDL-cholesterol (HDL-C), non-HDL-C, and triglycerides (TG). RESULTS In this Yup'ik study population, we found that up to 21.6% of the total effects of adiposity on plasma TG, HDL-C, and non-HDL-C are mediated through measures of insulin resistance or sensitivity. Moreover, RBC DHA and EPA moderated the positive association between waist circumference (WC) and TC or non-HDL-C, whereas only DHA moderated the positive association between WC and TG. However, the indirect path between WC and plasma lipids was not significantly moderated by dietary n-3 PUFAs. CONCLUSIONS Intake of n-3 PUFAs may independently reduce dyslipidemia through the direct path resulting from excess adiposity in Yup'ik adults. NIR moderation effects suggest that additional nutrients contained in n-3 PUFA-rich foods may also reduce dyslipidemia.
Collapse
Affiliation(s)
- Bert B Boyer
- Oregon Health & Science University, Departments of Obstetrics and Gynecology and Medicine, Portland, OR, USA.
| | - Howard W Wiener
- University of Alabama at Birmingham, Department of Epidemiology, Birmingham, AL, USA
| | - Scarlett E Hopkins
- Oregon Health & Science University, Departments of Obstetrics and Gynecology and Medicine, Portland, OR, USA
| | - Jonathan Q Purnell
- Oregon Health & Science University, Departments of Obstetrics and Gynecology and Medicine, Portland, OR, USA
| | - Diane M O'Brien
- University of Alaska Fairbanks, Department of Biology and Wildlife, Fairbanks, AK, USA
| | - Theresa Aliwarga
- University of Washington, Department of Pharmaceutics, Seattle, WA, USA
| | - Jeremy J Pomeroy
- Marshfield Clinic, Clinical Research Center, Marshfield, WI, USA
| | - Joseph E Aslan
- Oregon Health & Science University, Departments of Obstetrics and Gynecology and Medicine, Portland, OR, USA
| | - Kenneth E Thummel
- University of Washington, Department of Pharmaceutics, Seattle, WA, USA
| | - Hemant K Tiwari
- University of Alabama at Birmingham, Department of Biostatistics, Birmingham, AL, USA
| |
Collapse
|
46
|
Zhu Y, Zhang J, Wang C, Zheng T, Di S, Wang Y, Fei W, Liang W, Wang L. Ameliorative Effect of Ethanolic Echinacea purpurea against Hyperthyroidism-Induced Oxidative Stress via AMRK and PPAR Signal Pathway Using Transcriptomics and Network Pharmacology Analysis. Int J Mol Sci 2022; 24:ijms24010187. [PMID: 36613632 PMCID: PMC9820381 DOI: 10.3390/ijms24010187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022] Open
Abstract
Echinacea purpurea (L.) Moench (EP) is a well-known botanical supplement with antioxidant characteristics. However, the effects of EP on oxidative stress induced by hyperthyroidism have not yet been studied. This study was designed to evaluate the antioxidative effect of ethanolic Echinacea Purpurea (EEP) on hyperthyroidism-induced oxidative stress mice using an integrated strategy combining transcriptomics with network pharmacology analysis. Firstly, a hyperthyroidism mice model was induced via thyroxine (160 mg/kg) and EEP (1, 2, or 4 g/kg) once daily for 2 weeks. Body weight, thyroid-stimulating hormones, and oxidative stress markers were tested. Secondly, EEP regulating the potential genes at transcript level were analyzed. Thirdly, a network pharmacology based on the constituents of EEP identified using UPLC-Q-TOF-MS analysis was adopted. Finally, a joint analysis was performed to identify the key pathway. The results showed that EEP significantly changed the thyroid-stimulating hormones and oxidative stress markers. Meanwhile, RT-qPCR and Western Blotting demonstrated that the mechanism of the antioxidant effect of EEP reversed the mRNA expression of EHHADH, HMGCR and SLC27A2 and the protein expression of FABP and HMGCR in AMPK and PPAR signaling pathways. This study integrates transcriptomics with network pharmacology to reveal the mechanism of ameliorative effect of EEP on hyperthyroidism-induced oxidative stress.
Collapse
Affiliation(s)
- Yingli Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- Correspondence: (J.Z.); (L.W.)
| | - Chun Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ting Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Songrui Di
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yinyin Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenting Fei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weican Liang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
- Correspondence: (J.Z.); (L.W.)
| |
Collapse
|
47
|
Zeng W, Yin X, Jiang Y, Jin L, Liang W. PPARα at the crossroad of metabolic-immune regulation in cancer. FEBS J 2022; 289:7726-7739. [PMID: 34480827 DOI: 10.1111/febs.16181] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/04/2021] [Accepted: 09/03/2021] [Indexed: 01/14/2023]
Abstract
Rewiring metabolism to sustain cell growth, division, and survival is the most prominent feature of cancer cells. In particular, dysregulated lipid metabolism in cancer has received accumulating interest, since lipid molecules serve as cell membrane structure components, secondary signaling messengers, and energy sources. Given the critical role of immune cells in host defense against cancer, recent studies have revealed that immune cells compete for nutrients with cancer cells in the tumor microenvironment and accordingly develop adaptive metabolic strategies for survival at the expense of compromised immune functions. Among these strategies, lipid metabolism reprogramming toward fatty acid oxidation is closely related to the immunosuppressive phenotype of tumor-infiltrated immune cells, including macrophages and dendritic cells. Therefore, it is important to understand the lipid-mediated crosstalk between cancer cells and immune cells in the tumor microenvironment. Peroxisome proliferator-activated receptors (PPARs) consist of a nuclear receptor family for lipid sensing, and one of the family members PPARα is responsible for fatty acid oxidation, energy homeostasis, and regulation of immune cell functions. In this review, we discuss the emerging role of PPARα-associated metabolic-immune regulation in tumor-infiltrated immune cells, and key metabolic events and pathways involved, as well as their influences on antitumor immunity.
Collapse
Affiliation(s)
- Wenfeng Zeng
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaozhe Yin
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Yunhan Jiang
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lingtao Jin
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Wei Liang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
48
|
Panyard DJ, Yu B, Snyder MP. The metabolomics of human aging: Advances, challenges, and opportunities. SCIENCE ADVANCES 2022; 8:eadd6155. [PMID: 36260671 PMCID: PMC9581477 DOI: 10.1126/sciadv.add6155] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/01/2022] [Indexed: 05/02/2023]
Abstract
As the global population becomes older, understanding the impact of aging on health and disease becomes paramount. Recent advancements in multiomic technology have allowed for the high-throughput molecular characterization of aging at the population level. Metabolomics studies that analyze the small molecules in the body can provide biological information across a diversity of aging processes. Here, we review the growing body of population-scale metabolomics research on aging in humans, identifying the major trends in the field, implicated biological pathways, and how these pathways relate to health and aging. We conclude by assessing the main challenges in the research to date, opportunities for advancing the field, and the outlook for precision health applications.
Collapse
Affiliation(s)
- Daniel J. Panyard
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
49
|
Wang D, Ali F, Liu H, Cheng Y, Wu M, Saleem MZ, Zheng H, Wei L, Chu J, Xie Q, Shen A, Peng J. Quercetin inhibits angiotensin II-induced vascular smooth muscle cell proliferation and activation of JAK2/STAT3 pathway: A target based networking pharmacology approach. Front Pharmacol 2022; 13:1002363. [PMID: 36324691 PMCID: PMC9618806 DOI: 10.3389/fphar.2022.1002363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Abstract
The rapid growth of vascular smooth muscle cells (VSMCs) represents crucial pathological changes during the development of hypertensive vascular remodeling. Although quercetin exhibits significantly therapeutic effects on antihypertension, the systematic role of quercetin and its exact mode of action in relation to the VSMCs growth and its hypertension-related networking pharmacology is not well-documented. Therefore, the effect of quercetin was investigated using networking pharmacology followed by in vitro strategies to explore its efficacy against angiotensin II (Ang II)-induced cell proliferation. Putative genes of hypertension and quercetin were collected using database mining, and their correlation was investigated. Subsequently, a network of protein-protein interactions was constructed and gene ontology (GO) analysis was performed to identify the role of important genes (including CCND1) and key signaling pathways [including cell proliferation and Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway]. We therefore further investigated the effects of quercetin in Ang II-stimulated VSMCs. This current research revealed that quercetin significantly reduced the cell confluency, cell number, and cell viability, as well as expression of proliferating cell nuclear antigen (PCNA) in Ang II-stimulated VSMCs. Mechanistic study by western blotting confirmed that quercetin treatment attenuated the activation of JAK2 and STAT3 by reducing its phosphorylation in Ang II stimulated VSMCs. Collectively, the current study revealed the inhibitory effects of quercetin on proliferation of Ang II stimulated VSMCs, by inhibiting the activation of JAK2/STAT3 signaling might be one of underlying mechanisms.
Collapse
Affiliation(s)
- Di Wang
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Farman Ali
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Huixin Liu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Ying Cheng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Meizhu Wu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Muhammad Zubair Saleem
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Huifang Zheng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Lihui Wei
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Jiangfeng Chu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Qiurong Xie
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Aling Shen
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- *Correspondence: Aling Shen, ; Jun Peng,
| | - Jun Peng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- *Correspondence: Aling Shen, ; Jun Peng,
| |
Collapse
|
50
|
Halim NFAA, Ali MSM, Leow ATC, Rahman RNZRA. Membrane fatty acid desaturase: biosynthesis, mechanism, and architecture. Appl Microbiol Biotechnol 2022; 106:5957-5972. [PMID: 36063178 DOI: 10.1007/s00253-022-12142-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022]
Abstract
Fatty acid desaturase catalyzes the desaturation reactions by inserting double bonds into the fatty acyl chain, producing unsaturated fatty acids, which play a vital part in the synthesis of polyunsaturated fatty acids. Though soluble fatty acid desaturases have been described extensively in advanced organisms, there are very limited studies of membrane fatty acid desaturases due to their difficulties in producing a sufficient amount of recombinant desaturases. However, the advancement of technology has shown substantial progress towards the development of elucidating crystal structures of membrane fatty acid desaturase, thus, allowing modification of structure to be manipulated. Understanding the structure, mechanism, and biosynthesis of fatty acid desaturase lay a foundation for the potential production of various strategies associated with alteration and modifications of polyunsaturated fatty acids. This manuscript presents the current state of knowledge and understanding about the structure, mechanisms, and biosynthesis of fatty acid desaturase. In addition, the role of unsaturated fatty acid desaturases in health and diseases is also encompassed. This will be useful in understanding the molecular basis and structural protein of fatty acid desaturase that are significant for the advancement of therapeutic strategies associated with the improvement of health status. KEY POINTS: • Current state of knowledge and understanding about the biosynthesis, mechanisms, and structure of fatty acid desaturase. • The role of unsaturated fatty acid desaturase. • The molecular basis and structural protein elucidated the crystal structure of fatty acid desaturase.
Collapse
Affiliation(s)
- Nur Farah Anis Abd Halim
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohd Shukuri Mohamad Ali
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Adam Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Raja Noor Zaliha Raja Abd Rahman
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|